Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28.324
Filtrar
Más filtros

Intervalo de año de publicación
1.
Front Immunol ; 15: 1427554, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39114662

RESUMEN

Inflammatory myofibroblastic tumor (IMT) is a rare pathological entity first described in 1939. This lesion is most commonly found in the lungs, but cases involving other systems, such as the central nervous system known as intracranial IMT (IIMT), have also been reported. Diagnosis currently relies on pathological results due to the lack of characteristic imaging changes. Surgical resection is an effective treatment, though the disease is invasive and may recur. Previous literature has reported a high level of programmed death 1 (PD-1) expression in IMT tissues, suggesting that immunotherapy may be effective for this condition. In this case report, we present a middle-aged male who received PD-1 inhibitor and oncolytic adenovirus (Ad-TD-nsIL12) treatment after IIMT resection surgery. This successful approach provides a new direction for the treatment of IIMT.


Asunto(s)
Adenoviridae , Neoplasias Encefálicas , Inhibidores de Puntos de Control Inmunológico , Viroterapia Oncolítica , Humanos , Masculino , Viroterapia Oncolítica/métodos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Encefálicas/terapia , Persona de Mediana Edad , Adenoviridae/genética , Virus Oncolíticos/genética , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias de Tejido Muscular/terapia , Terapia Combinada , Resultado del Tratamiento
2.
Int J Mol Sci ; 25(15)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39126087

RESUMEN

Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Vacunas Virales , Animales , Cricetinae , Vacunas Virales/inmunología , Marburgvirus/inmunología , Enfermedad del Virus de Marburg/prevención & control , Enfermedad del Virus de Marburg/inmunología , Modelos Animales de Enfermedad , Adenoviridae/genética , Adenoviridae/inmunología , Vesiculovirus/inmunología , Vesiculovirus/genética , Anticuerpos Antivirales/inmunología , Vacunación/métodos
3.
Front Immunol ; 15: 1376395, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975350

RESUMEN

Influenza A Virus (IAV) and Respiratory Syncytial Virus (RSV) are both responsible for millions of severe respiratory tract infections every year worldwide. Effective vaccines able to prevent transmission and severe disease, are important measures to reduce the burden for the global health system. Despite the strong systemic immune responses induced upon current parental immunizations, this vaccination strategy fails to promote a robust mucosal immune response. Here, we investigated the immunogenicity and efficacy of a mucosal adenoviral vector vaccine to tackle both pathogens simultaneously at their entry site. For this purpose, BALB/c mice were immunized intranasally with adenoviral vectors (Ad) encoding the influenza-derived proteins, hemagglutinin (HA) and nucleoprotein (NP), in combination with an Ad encoding for the RSV fusion (F) protein. The mucosal combinatory vaccine induced neutralizing antibodies as well as local IgA responses against both viruses. Moreover, the vaccine elicited pulmonary CD8+ and CD4+ tissue resident memory T cells (TRM) against the immunodominant epitopes of RSV-F and IAV-NP. Furthermore, the addition of Ad-TGFß or Ad-CCL17 as mucosal adjuvant enhanced the formation of functional CD8+ TRM responses against the conserved IAV-NP. Consequently, the combinatory vaccine not only provided protection against subsequent infections with RSV, but also against heterosubtypic challenges with pH1N1 or H3N2 strains. In conclusion, we present here a potent combinatory vaccine for mucosal applications, which provides protection against two of the most relevant respiratory viruses.


Asunto(s)
Anticuerpos Antivirales , Inmunidad Mucosa , Virus de la Influenza A , Vacunas contra la Influenza , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Animales , Ratones , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Anticuerpos Antivirales/inmunología , Virus de la Influenza A/inmunología , Femenino , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Virus Sincitiales Respiratorios/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Vacunas Combinadas/inmunología , Vacunas Combinadas/administración & dosificación , Humanos , Adenoviridae/inmunología , Adenoviridae/genética , Vectores Genéticos
4.
ACS Nano ; 18(28): 18282-18298, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38953884

RESUMEN

The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica , Virus Oncolíticos , Replicación Viral , Animales , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/fisiología , Virus Oncolíticos/fisiología , Virus Oncolíticos/genética , Replicación Viral/efectos de la radiación , Ratones , Humanos , Línea Celular Tumoral , Terapia por Ultrasonido/métodos , Melanoma/terapia , Melanoma/patología
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000138

RESUMEN

The ongoing battle against viral pandemics continues, with the possibility of future outbreaks. The search for effective antiviral compounds that can combat a diverse range of viruses continues to be a focal point of research. This study investigated the efficacy of two natural antimicrobial peptides (AMPs) (lactoferricin and LL-37), two synthetic AMPs (melimine and Mel4), and nine AMP mimics (758, 1091, 1096, 1083, 610, NAPL, 3-BIPL, 4-BIPL, and Sau-22) against influenza A virus strains H1N1 and H3N2, human adenovirus 5 (HAdV-5), and murine norovirus 1 (MNV-1). These compounds were tested using virus pre-treatment, cell pre-treatment, or post-cell entry treatment assays, electron microscopy, and circular dichroism (CD), alongside evaluations of cytotoxicity against the host cells. After virus pre-treatment, the AMP mimics 610 and Sau-22 had relatively low IC50 values for influenza strains H1N1 (2.35 and 6.93 µM, respectively) and H3N2 (3.7 and 5.34 µM, respectively). Conversely, natural and synthetic AMPs were not active against these strains. For the non-enveloped viruses, the AMP Mel4 and mimic 1083 had moderate activity against HAdV-5 (Mel4 IC50 = 47.4 µM; 1083 IC50 = 47.2 µM), whereas all AMPs, but none of the mimics, were active against norovirus (LL-37 IC50 = 4.2 µM; lactoferricin IC50 = 23.18 µM; melimine IC50 = 4.8 µM; Mel4 IC50 = 8.6 µM). Transmission electron microscopy demonstrated that the mimics targeted the outer envelope of influenza viruses, while the AMPs targeted the capsid of non-enveloped viruses. CD showed that Mel4 adopted an α-helical structure in a membrane mimetic environment, but mimic 758 remained unstructured. The diverse activity against different virus groups is probably influenced by charge, hydrophobicity, size, and, in the case of natural and synthetic AMPs, their secondary structure. These findings underscore the potential of peptides and mimics as promising candidates for antiviral therapeutics against both enveloped and non-enveloped viruses.


Asunto(s)
Antivirales , Norovirus , Norovirus/efectos de los fármacos , Animales , Humanos , Ratones , Antivirales/farmacología , Antivirales/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Perros , Adenoviridae/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Células de Riñón Canino Madin Darby , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/química
6.
mSystems ; 9(7): e0050524, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38953320

RESUMEN

Nanopore direct RNA sequencing (DRS) enables the capture and full-length sequencing of native RNAs, without recoding or amplification bias. Resulting data sets may be interrogated to define the identity and location of chemically modified ribonucleotides, as well as the length of poly(A) tails, on individual RNA molecules. The success of these analyses is highly dependent on the provision of high-resolution transcriptome annotations in combination with workflows that minimize misalignments and other analysis artifacts. Existing software solutions for generating high-resolution transcriptome annotations are poorly suited to small gene-dense genomes of viruses due to the challenge of identifying distinct transcript isoforms where alternative splicing and overlapping RNAs are prevalent. To resolve this, we identified key characteristics of DRS data sets that inform resulting read alignments and developed the nanopore guided annotation of transcriptome architectures (NAGATA) software package (https://github.com/DepledgeLab/NAGATA). We demonstrate, using a combination of synthetic and original DRS data sets derived from adenoviruses, herpesviruses, coronaviruses, and human cells, that NAGATA outperforms existing transcriptome annotation software and yields a consistently high level of precision and recall when reconstructing both gene sparse and gene-dense transcriptomes. Finally, we apply NAGATA to generate the first high-resolution transcriptome annotation of the neglected pathogen human adenovirus type F41 (HAdV-41) for which we identify 77 distinct transcripts encoding at least 23 different proteins. IMPORTANCE: The transcriptome of an organism denotes the full repertoire of encoded RNAs that may be expressed. This is critical to understanding the biology of an organism and for accurate transcriptomic and epitranscriptomic-based analyses. Annotating transcriptomes remains a complex task, particularly in small gene-dense organisms such as viruses which maximize their coding capacity through overlapping RNAs. To resolve this, we have developed a new software nanopore guided annotation of transcriptome architectures (NAGATA) which utilizes nanopore direct RNA sequencing (DRS) datasets to rapidly produce high-resolution transcriptome annotations for diverse viruses and other organisms.


Asunto(s)
Anotación de Secuencia Molecular , Programas Informáticos , Transcriptoma , Humanos , Transcriptoma/genética , Anotación de Secuencia Molecular/métodos , Análisis de Secuencia de ARN/métodos , Herpesviridae/genética , Coronavirus/genética , Secuenciación de Nanoporos/métodos , Nanoporos , Adenoviridae/genética
7.
J Virol ; 98(7): e0062224, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38953377

RESUMEN

African swine fever virus causes a lethal hemorrhagic disease in domestic swine and wild boar for which currently licensed commercial vaccines are only available in Vietnam. Development of subunit vaccines is complicated by the lack of information on protective antigens as well as suitable delivery systems. Our previous work showed that a pool of eight African swine fever virus genes vectored using an adenovirus prime and modified vaccinia virus boost could prevent fatal disease after challenge with a virulent genotype I isolate of the virus. Here, we identify antigens within this pool of eight that are essential for the observed protection and demonstrate that adenovirus-prime followed by adenovirus-boost can also induce protective immune responses against genotype I African swine fever virus. Immunization with a pool of adenoviruses expressing individual African swine fever virus genes partially tailored to genotype II virus did not protect against challenge with genotype II Georgia 2007/1 strain, suggesting that different antigens may be required to induce cross-protection for genetically distinct viruses. IMPORTANCE: African swine fever virus causes a lethal hemorrhagic disease in domestic pigs and has killed millions of animals across Europe and Asia since 2007. Development of safe and effective subunit vaccines against African swine fever has been problematic due to the complexity of the virus and a poor understanding of protective immunity. In a previous study, we demonstrated that a complex combination of eight different virus genes delivered using two different viral vector vaccine platforms protected domestic pigs from fatal disease. In this study, we show that three of the eight genes are required for protection and that one viral vector is sufficient, significantly reducing the complexity of the vaccine. Unfortunately, this combination did not protect against the current outbreak strain of African swine fever virus, suggesting that more work to identify immunogenic and protective viral proteins is required to develop a truly effective African swine fever vaccine.


Asunto(s)
Adenoviridae , Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Vectores Genéticos , Genotipo , Vacunas Virales , Animales , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/prevención & control , Fiebre Porcina Africana/virología , Fiebre Porcina Africana/inmunología , Porcinos , Vacunas Virales/inmunología , Vacunas Virales/genética , Vacunas Virales/administración & dosificación , Vectores Genéticos/genética , Adenoviridae/genética , Adenoviridae/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/genética , Antígenos Virales/inmunología , Antígenos Virales/genética
8.
Signal Transduct Target Ther ; 9(1): 190, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39039046

RESUMEN

The upper respiratory tract is the initial site of SARS-CoV-2 infection. Nasal spike-specific secretory immunoglobulin A (sIgA) correlates with protection against Omicron breakthrough infection. We report that intranasal vaccination using human adenovirus serotype 5 (Ad5) vectored Omicron spike in people who previously vaccinated with ancestral vaccine could induce robust neutralizing sIgA in the nasal passage. Nasal sIgA was predominantly present in dimeric and multimeric forms and accounted for nearly 40% of total proteins in nasal mucosal lining fluids (NMLFs). A low-level IgG could also be detected in NMLFs but not IgM, IgD, and IgE. After a complete nasal wash, sIgA in the nasal passage could be replenished rapidly within a few hours. A comparison of purified paired serum IgA, serum IgG, and nasal sIgA from the same individuals showed that sIgA was up to 3-logs more potent than serum antibodies in binding to spikes and in neutralizing Omicron subvariants. Serum IgG and IgA failed to neutralize XBB and BA.2.86, while nasal sIgA retained potent neutralization against these newly emerged variants. Further analysis showed that sIgA was more effective than IgG or IgA in blocking spike-mediated cell-to-cell transmission and protecting hACE2 mice from XBB challenge. Using a sIgA monoclonal antibody as a reference, we estimated that the total nasal sIgA contains about 2.6-3.9% spike-specific sIgA in NMLFs collected approximately one month after intranasal vaccination. Our study provided insights for developing intranasal vaccines that can induce sIgA to build an effective and mutation-resistant first-line immune barrier against constantly emerging variants.


Asunto(s)
Administración Intranasal , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Animales , Ratones , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/genética , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Femenino , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Inmunoglobulina A Secretora/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Masculino
9.
J Microbiol ; 62(7): 491-509, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39037484

RESUMEN

Adenoviral vectors are crucial for gene therapy and vaccine development, offering a platform for gene delivery into host cells. Since the discovery of adenoviruses, first-generation vectors with limited capacity have evolved to third-generation vectors flacking viral coding sequences, balancing safety and gene-carrying capacity. The applications of adenoviral vectors for gene therapy and anti-viral treatments have expanded through the use of in vitro ligation and homologous recombination, along with gene editing advancements such as CRISPR-Cas9. Current research aims to maintain the efficacy and safety of adenoviral vectors by addressing challenges such as pre-existing immunity against adenoviral vectors and developing new adenoviral vectors from rare adenovirus types and non-human species. In summary, adenoviral vectors have great potential in gene therapy and vaccine development. Through continuous research and technological advancements, these vectors are expected to lead to the development of safer and more effective treatments.


Asunto(s)
Adenoviridae , Edición Génica , Terapia Genética , Vectores Genéticos , Vectores Genéticos/genética , Humanos , Adenoviridae/genética , Terapia Genética/métodos , Edición Génica/métodos , Animales , Sistemas CRISPR-Cas , Técnicas de Transferencia de Gen , Desarrollo de Vacunas
10.
Viruses ; 16(7)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-39066180

RESUMEN

Since the significance of viral infections in children and adolescents with nephrotic syndrome (NS) is yet to be defined, this study intended to estimate the occurrence, pattern, and outcomes of some DNA viral infections in children with NS. METHODS: A prospective study was conducted to determine the genome identification of the viruses Epstein-Barr (EBV), human cytomegalovirus (HCMV), human herpesvirus 6 (HHV-6 type A and type B) and 7 (HHV-7), polyomavirus (BKV), and human adenovirus (HAdV) in plasma and urine samples of pediatric patients with NS. RESULTS: A total of 35 patients aged 1 to 18 years with NS and under immunosuppressant drugs participated in the study. Plasma and urine samples were collected at regular intervals during a median follow-up of 266 days (range 133-595), and DNA was analyzed to detect the selected DNA viruses. Eleven patients (31.4%) had active virus infections, and patterns were classified as coinfection, recurrent, and consecutive. Of these, six patients (54.5%) presented viral coinfection, six (54.5%) viral recurrence, and seven patients (63.3%) had viral consecutive infection. Ten of the eleven patients with active infection had a proteinuria relapse (91%) and eight (72.7%) were hospitalized (p = 0.0022). Active HCMV infection was the most frequent infection and was observed in six patients (54.5%), three of the eleven patients (27.2%) had suspected HCMV disease in the gastrointestinal tract, and one had HHV-7 coinfection. The frequency of other infections was: 9% for HHV-6, 45.5% for BKV, 27.3% for HHV-7, 18.2% for EBV, and 18.2% for HAdV. CONCLUSION: viral infections, especially HCMV, can be an important cause of morbidity and nephrotic syndrome relapse in children.


Asunto(s)
Virus BK , Síndrome Nefrótico , Humanos , Síndrome Nefrótico/virología , Síndrome Nefrótico/complicaciones , Adolescente , Niño , Masculino , Femenino , Preescolar , Virus BK/genética , Virus BK/aislamiento & purificación , Lactante , Estudios Prospectivos , ADN Viral/genética , Herpesviridae/genética , Herpesviridae/clasificación , Herpesviridae/aislamiento & purificación , Coinfección/virología , Infecciones por Herpesviridae/virología , Adenoviridae/genética , Adenoviridae/aislamiento & purificación , Adenoviridae/clasificación
11.
Viruses ; 16(7)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-39066256

RESUMEN

Adenoviruses are non-enveloped DNA viruses that cause a wide range of symptoms, from mild infections to life-threatening diseases in a broad range of hosts. Due to the unique characteristics of these viruses, they have also become a vehicle for gene-transfer and cancer therapeutic instruments. Adenovirus vectors can be used in gene therapy by modifying wild-type viruses to render them replication-defective. This makes it possible to swap out particular viral genes for segments that carry therapeutic genes and to employ the resultant vector as a means of delivering genes to specified tissues. In this review, we outline the progressive development of adenovirus vectors, exploring their characteristics, genetic modifications, and range of uses in clinical and preclinical settings. A significant emphasis is placed on their crucial role in advancing gene therapy, cancer therapy, immunotherapy, and the latest breakthroughs in vaccine development for various diseases.


Asunto(s)
Adenoviridae , Terapia Genética , Vectores Genéticos , Neoplasias , Humanos , Terapia Genética/métodos , Adenoviridae/genética , Vectores Genéticos/genética , Neoplasias/terapia , Neoplasias/virología , Animales , Inmunoterapia/métodos , Técnicas de Transferencia de Gen
12.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(6): 905-909, 2024 Jun 06.
Artículo en Chino | MEDLINE | ID: mdl-38955740

RESUMEN

To investigate the status and epidemiological characteristics of respiratory pathogens infections in children with influenza-like illnesses (ILI) in Beijing Children's Hospital from 2022 to 2023. A dual amplification technique was used to detect nucleic acids of seven common respiratory pathogens, including influenza A virus (Flu A), influenza B virus (Flu B), mycoplasma pneumoniae (MP), respiratory syncytial virus (RSV), parainfluenza virus (PIV), adenovirus (ADV), and Chlamydia pneumoniae (CP), in outpatient and inpatient children (aged 0-18 years) with influenza-like symptoms who sought medical care at Beijing Children's Hospital, from January 2022 to March 2023. A total of 43 663 children were included in the study, of which 27 903 tested positive for respiratory pathogens with a total detection rate of 63.91%. Flu A had the highest detection rate of 69.93% (27 332/39 084), followed by MP about 13.22% (380/2 875). The total detection rate of RSV, PIV and ADV was 7.69% (131/1 704). Flu B had a detection rate of 0.16% (64/39 084). No CP was detected in this study. A total of 7 cases of dual infections were detected, with a detection rate of 0.41% (7/1 704). The Chi-square test was used to analyze the differences in detection rates of pathogens among different genders, age groups, and different seasons. Among the seven pathogens, only Flu A had statistically significant differences in gender (χ2=16.712, P<0.001). The detection rates of Flu A and MP showed an increasing trend with age (both P trend<0.001), while the detection rates of RSV and PIV showed a decreasing trend with age (both P trend<0.001). Flu A had its epidemic peak in winter and spring, with detection rates of 61.30% (3 907/6 374) and 77.47% (23 207/29 958) respectively; MP and PIV had higher detection rates in autumn (25.14% and 7.64% respectively); RSV showed a relatively higher detection rate in winter (8.69%); Flu B and ADV had lower detection rates throughout the study period (0.16% and 1.17% respectively). In conclusion, children with ILI in 2022-2023 were mainly infected with a single respiratory pathogen, and occasionally dual pathogen infections were observed. Among them, the detection rate of Flu A was the highest, and only Flu A showed a gender difference in detection rate. As the age of the children patients increased, the detection rate of Flu A and MP showed an increasing trend, while RSV and PIV showed a decreasing trend. The prevalence of Flu A, Flu B, MP, PIV, and RSV were seasonal.


Asunto(s)
Gripe Humana , Infecciones del Sistema Respiratorio , Humanos , Niño , Preescolar , Lactante , Adolescente , Gripe Humana/epidemiología , Masculino , Femenino , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/microbiología , Beijing/epidemiología , Virus de la Influenza B/aislamiento & purificación , Virus de la Influenza A/aislamiento & purificación , Mycoplasma pneumoniae/aislamiento & purificación , Recién Nacido , Virus Sincitiales Respiratorios/aislamiento & purificación , Hospitales Pediátricos , Chlamydophila pneumoniae/aislamiento & purificación , Infecciones por Virus Sincitial Respiratorio/epidemiología , China/epidemiología , Adenoviridae/aislamiento & purificación
13.
Front Immunol ; 15: 1355566, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38835775

RESUMEN

Dendritic cell (DC)-based vaccines have emerged as a promising strategy in cancer immunotherapy due to low toxicity. However, the therapeutic efficacy of DC as a monotherapy is insufficient due to highly immunosuppressive tumor environment. To address these limitations of DC as immunotherapeutic agent, we have developed a polymeric nanocomplex incorporating (1) oncolytic adenovirus (oAd) co-expressing interleukin (IL)-12 and granulocyte-macrophage colony-stimulating factor (GM-CSF) and (2) arginine-grafted bioreducible polymer with PEGylated paclitaxel (APP) to restore antitumor immune surveillance function in tumor milieu and potentiate immunostimulatory attributes of DC vaccine. Nanohybrid complex (oAd/APP) in combination with DC (oAd/APP+DC) induced superior expression level of antitumor cytokines (IL-12, GM-CSF, and interferon gamma) than either oAd/APP or DC monotherapy in tumor tissues, thus resulting in superior intratumoral infiltration of both endogenous and exogenous DCs. Furthermore, oAd/APP+DC treatment led superior migration of DC to secondary lymphoid organs, such as draining lymph nodes and spleen, in comparison with either monotherapy. Superior migration profile of DCs in oAd/APP+DC treatment group resulted in more prolific activation of tumor-specific T cells in these lymphoid organs and greater intratumoral infiltration of T cells. Additionally, oAd/APP+DC treatment led to lower subset of tumor infiltrating lymphocytes and splenocytes being immunosuppressive regulatory T cells than any other treatment groups. Collectively, oAd/APP+DC led to superior induction of antitumor immune response and amelioration of immunosuppressive tumor microenvironment to elicit potent tumor growth inhibition than either monotherapy.


Asunto(s)
Adenoviridae , Células Dendríticas , Viroterapia Oncolítica , Virus Oncolíticos , Paclitaxel , Células Dendríticas/inmunología , Animales , Paclitaxel/farmacología , Adenoviridae/genética , Ratones , Virus Oncolíticos/inmunología , Virus Oncolíticos/genética , Viroterapia Oncolítica/métodos , Terapia Combinada , Línea Celular Tumoral , Humanos , Ratones Endogámicos C57BL , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Femenino , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos
14.
Sci Rep ; 14(1): 13108, 2024 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849383

RESUMEN

LHPP has been shown to be a new tumor suppressor, and has a tendency to be under-expressed in a variety of cancers. Oncolytic virotheray is a promising therapeutics for lung cancer in recent decade years. Here we successfully constructed a new recombinant oncolytic adenovirus GD55-LHPP and investigated the effect of GD55-LHPP on the growth of lung cancer cells in vitro and in vivo. The results showed that LHPP had lower expression in either lung cancer cells or clinical lung cancer tissues compared with normal cells or tissues, and GD55-LHPP effectively mediated LHPP expression in lung cancer cells. GD55-LHPP could effectively inhibit the proliferation of lung cancer cell lines and rarely affected normal cell growth. Mechanically, the oncolytic adenovirus GD55-LHPP was able to induce stronger apoptosis of lung cancer cells compared with GD55 through the activation of caspase signal pathway. Notably, GD55-LHPP also activated autophagy-related signal pathway. Further, GD55-LHPP efficiently inhibited tumor growth in lung cancer xenograft in mice and prolonged animal survival rate compared with the control GD55 or PBS. In conclusion, the novel construct GD55-LHPP provides a valuable strategy for lung cancer-targeted therapy and develop the role of tumor suppress gene LHPP in lung cancer gene therapy.


Asunto(s)
Adenoviridae , Apoptosis , Neoplasias Pulmonares , Viroterapia Oncolítica , Virus Oncolíticos , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Humanos , Animales , Viroterapia Oncolítica/métodos , Adenoviridae/genética , Virus Oncolíticos/genética , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Desnudos , Femenino , Autofagia
15.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928259

RESUMEN

Oncolytic adenoviruses are in development as immunotherapeutic agents for solid tumors. Their efficacy is in part dependent on their ability to replicate in tumors. It is, however, difficult to obtain evidence for intratumoral oncolytic adenovirus replication if direct access to the tumor is not possible. Detection of systemic adenovirus DNA, which is sometimes used as a proxy, has limited value because it does not distinguish between the product of intratumoral replication and injected virus that did not replicate. Therefore, we investigated if detection of virus-associated RNA (VA RNA) by RT-qPCR on liquid biopsies could be used as an alternative. We found that VA RNA is expressed in adenovirus-infected cells in a replication-dependent manner and is secreted by these cells in association with extracellular vesicles. This allowed VA RNA detection in the peripheral blood of a preclinical in vivo model carrying adenovirus-injected human tumors and on liquid biopsies from a human clinical trial. Our results confirm that VA RNA detection in liquid biopsies can be used for minimally invasive assessment of oncolytic adenovirus replication in solid tumors in vivo.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica , Virus Oncolíticos , ARN Viral , Replicación Viral , Humanos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , ARN Viral/genética , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Viroterapia Oncolítica/métodos , Ratones , Línea Celular Tumoral , Neoplasias/terapia , Neoplasias/genética , Femenino
16.
Nano Lett ; 24(31): 9598-9607, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38922640

RESUMEN

Enhancing the antitumor immune response and targeting ability of oncolytic viruses will improve the effect of tumor immunotherapy. Through infecting neural stem cells (NSCs) with a capsid dual-modified oncolytic adenovirus (CRAd), we obtained and characterized the "oncolytic extracellular vesicles" (CRAdEV) with improved targeted infection and tumor killing activity compared with CRAd. Both ex vivo and in vivo studies revealed that CRAdEV activated innate immune cells and importantly enhanced the immunomodulatory effect compared to CRAd. We found that CRAdEV effectively increased the number of DCs and activated CD4+ and CD8+ T cells, significantly increased the number and activation of B cells, and produced higher levels of tumor-specific antibodies, thus eliciting enhanced antitumor activity compared with CRAd in a B16 xenograft immunocompetent mice model. This study provides a novel approach to oncolytic adenovirus modification and demonstrates the potential of "oncolytic extracellular vesicles" in antitumor immunotherapy.


Asunto(s)
Adenoviridae , Vesículas Extracelulares , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Ratones , Adenoviridae/genética , Viroterapia Oncolítica/métodos , Humanos , Línea Celular Tumoral , Inmunoterapia , Células-Madre Neurales/inmunología , Inmunomodulación/efectos de los fármacos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/patología , Linfocitos T CD8-positivos/inmunología
17.
Int Immunopharmacol ; 137: 112393, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38852522

RESUMEN

Recombinant adenovirus serotype 5 (Ad5)-mediated virotherapy is a maturing technique in cancer treatment. However, the utility of adenovirus (Ad) has been limited by low expression of coxsackievirus and adenovirus receptor (CAR) in cancer cells resulting in poor infectivity of Ads. To overcome the problem, we aimed to develop a novel tropism-modified oncolytic adenovirus, ZD55-F-HI-sPD-1-EGFP, which contains the epitope of PD-1 (70-77aa) at the HI-loop of Ad fiber. Trimerization of Fiber-sPD-1 was confirmed by immunoblot analysis. ZD55-F-HI-sPD-1-EGFP shows a remarkable improvement in viral infection rate and gene transduction efficiency in the PD-L1-positive cancer cells. Competition assays with a PD-L1 protein reveals that cell internalization of ZD55-F-HI-sPD-1-EGFP is mediated by both CAR and PD-L1 at a high dose. The progeny virus production capacity showed that sPD-1 incorporated fiber-modified oncolytic Ad replication was not affected. Furthermore, treating with ZD55-F-HI-sPD-1-EGFP significantly increased viral infection rate and enhanced anti-tumor effect in vivo. This study demonstrates that the strategy to expand tropism of oncolytic Ad may significantly improve therapeutic profile for cancer treatment.


Asunto(s)
Adenoviridae , Antígeno B7-H1 , Viroterapia Oncolítica , Virus Oncolíticos , Tropismo Viral , Humanos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Animales , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Línea Celular Tumoral , Ratones , Neoplasias/terapia , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Femenino , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Células HEK293
18.
Reprod Sci ; 31(8): 2199-2208, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38907125

RESUMEN

Fetal growth restriction (FGR) is associated with uteroplacental insufficiency, and neurodevelopmental and structural brain deficits in the infant. It is currently untreatable. We hypothesised that treating the maternal uterine artery with vascular endothelial growth factor adenoviral gene therapy (Ad.VEGF-A165) normalises offspring brain weight and prevents brain injury in a guinea pig model of FGR. Pregnant guinea pigs were fed a restricted diet before and after conception and received Ad.VEGF-A165 (1 × 1010 viral particles, n = 18) or vehicle (n = 18), delivered to the external surface of the uterine arteries, in mid-pregnancy. Pregnant, ad libitum-fed controls received vehicle only (n = 10). Offspring brain weight and histological indices of brain injury were assessed at term and 5-months postnatally. At term, maternal nutrient restriction reduced fetal brain weight and increased microglial ramification in all brain regions but did not alter indices of cell death, astrogliosis or myelination. Ad.VEGF-A165 increased brain weight and reduced microglial ramification in fetuses of nutrient restricted dams. In adult offspring, maternal nutrient restriction did not alter brain weight or markers of brain injury, whilst Ad.VEGF-A165 increased microglial ramification and astrogliosis in the hippocampus and thalamus, respectively. Ad.VEGF-A165 did not affect cell death or myelination in the fetal or offspring brain. Ad.VEGF-A165 normalises brain growth and markers of brain injury in guinea pig fetuses exposed to maternal nutrient restriction and may be a potential intervention to improve childhood neurodevelopmental outcomes in pregnancies complicated by FGR.


Asunto(s)
Adenoviridae , Encéfalo , Retardo del Crecimiento Fetal , Terapia Genética , Microglía , Arteria Uterina , Factor A de Crecimiento Endotelial Vascular , Animales , Cobayas , Embarazo , Femenino , Terapia Genética/métodos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Retardo del Crecimiento Fetal/terapia , Retardo del Crecimiento Fetal/metabolismo , Adenoviridae/genética , Encéfalo/metabolismo , Encéfalo/patología , Microglía/metabolismo , Desarrollo Fetal/fisiología , Vectores Genéticos
19.
Viruses ; 16(6)2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38932265

RESUMEN

Pathogenic adenovirus (Ad) infections are widespread but typically mild and transient, except in the immunocompromised. As vectors for gene therapy, vaccine, and oncology applications, Ad-based platforms offer advantages, including ease of genetic manipulation, scale of production, and well-established safety profiles, making them attractive tools for therapeutic development. However, the immune system often poses a significant challenge that must be overcome for adenovirus-based therapies to be truly efficacious. Both pre-existing anti-Ad immunity in the population as well as the rapid development of an immune response against engineered adenoviral vectors can have detrimental effects on the downstream impact of an adenovirus-based therapeutic. This review focuses on the different challenges posed, including pre-existing natural immunity and anti-vector immunity induced by a therapeutic, in the context of innate and adaptive immune responses. We summarise different approaches developed with the aim of tackling these problems, as well as their outcomes and potential future applications.


Asunto(s)
Inmunidad Adaptativa , Adenoviridae , Terapia Genética , Vectores Genéticos , Inmunidad Innata , Humanos , Adenoviridae/inmunología , Adenoviridae/genética , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Terapia Genética/métodos , Animales , Sistema Inmunológico/inmunología , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/terapia
20.
Transpl Infect Dis ; 26(4): e14296, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38830809

RESUMEN

BACKGROUND: Reactivation of viral infections, in particular cytomegalovirus (CMV) and adenovirus (ADV), cause morbidity and non-relapse-mortality in states of immune deficiency, especially after allogeneic hematopoietic cell transplantation (allo-HCT). Against the background of few available pharmacologic antiviral agents, limited by toxicities and resistance, adoptive transfer of virus-specific T-cells (VST) is a promising therapeutic approach. METHODS: We conducted a single-center retrospective analysis of adult patients treated with ADV- or CMV-specific T-cells in 2012-2022. Information was retrieved by review of electronic health records. Primary outcome was a response to VST by decreasing viral load or clinical improvement. Secondary outcomes included overall survival and safety of VST infusion, in particular association with graft-versus-host disease (GVHD). RESULTS: Ten patients were included, of whom four were treated for ADV, five for CMV, and one for ADV-CMV-coinfection. Cells were derived from stem cell donors (6/10) or third-party donors (4/10). Response criteria were met by six of 10 patients (4/4 ADV, 2/5 CMV, and 0/1 ADV-CMV). Overall survival was 40%. No infusion related adverse events were documented. Aggravation of GVHD after adoptive immunotherapy was observed in two cases, however in temporal association with a conventional donor lymphocyte infusion and a stem cell boost, respectively. CONCLUSION: In this cohort, CMV- and ADV-specific T-cell therapy appear to be safe and effective. We describe the first reported case of virus-specific T-cell therapy for CMV reactivation not associated with transplantation but with advanced HIV infection. This encourages further evaluation of adoptive immunotherapy beyond the context of allo-HCT.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Infecciones por VIH , Trasplante de Células Madre Hematopoyéticas , Linfocitos T , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Masculino , Estudios Retrospectivos , Femenino , Persona de Mediana Edad , Adulto , Citomegalovirus/inmunología , Linfocitos T/inmunología , Infecciones por VIH/inmunología , Enfermedad Injerto contra Huésped , Inmunoterapia Adoptiva/métodos , Carga Viral , Traslado Adoptivo/métodos , Adenoviridae/inmunología , Anciano , Resultado del Tratamiento , Infecciones por Adenoviridae/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA