Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 38(4): 573-594, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31828566

RESUMEN

The advent of immunotherapy has revolutionized how we manage and treat cancer. While the majority of immunotherapy-related studies performed to date have focused on adult malignancies, a handful of these therapies have also recently found success within the pediatric space. In this review, we examine the immunotherapeutic agents that have achieved the approval of the US Food and Drug Administration for treating childhood cancers, highlighting their development, mechanisms of action, and the lessons learned from the seminal clinical trials that ultimately led to their approval. We also shine a spotlight on several emerging immunotherapeutic modalities that we believe are poised to have a positive impact on the treatment of pediatric malignancies in the near future.


Asunto(s)
Inmunoterapia/tendencias , Neoplasias/inmunología , Neoplasias/terapia , Pediatría/tendencias , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Niño , Ensayos Clínicos Fase III como Asunto , Predicción , Humanos , Inmunoterapia/métodos , Pediatría/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto
2.
J Pediatr ; 201: 274-280.e12, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30041935

RESUMEN

We identified 13 patients with cat scratch (Bartonella henselae) bone infection among those admitted to a large tertiary care children's hospital over a 12-year period. The median age was 7 years and the median time from onset of illness to diagnosis was 10 days. Multifocal osteomyelitis involving spine and pelvis was common; no patient had a lytic bone lesion. Median treatment duration was 28 days (IQR, 24.5 days). Despite significant variations in treatment duration and antimicrobial therapy choices, all patients showed improvement.


Asunto(s)
Antibacterianos/uso terapéutico , Bartonella henselae/aislamiento & purificación , Enfermedad por Rasguño de Gato/complicaciones , Osteomielitis/etiología , Radiografía/métodos , Columna Vertebral/diagnóstico por imagen , Enfermedad por Rasguño de Gato/diagnóstico , Enfermedad por Rasguño de Gato/microbiología , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Osteomielitis/tratamiento farmacológico , Osteomielitis/microbiología , Estudios Retrospectivos
4.
Front Immunol ; 15: 1375413, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38895115

RESUMEN

Introduction: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas with unacceptably low cure rates occurring often in patients with neurofibromatosis 1 defects. To investigate oncolytic Herpes Simplex Virus (oHSV) as an immunotherapeutic approach, we compared viral replication, functional activity, and immune response between unarmed and interleukin 12 (IL-12)-armed oncolytic viruses in virus-permissive (B109) and -resistant (67C-4) murine MPNSTs. Methods: This study compared two attenuated IL-12-oHSVs with γ134.5 gene deletions (Δγ134.5) and the same transgene expression cassette. The primary difference in the IL-12-oHSVs was in their ability to counter the translational arrest response in infected cells. Unlike M002 (Δγ134.5, mIL-12), C002 (Δγ134.5, mIL-12, IRS1) expresses an HCMV IRS1 gene and evades dsRNA activated translational arrest in infected cells. Results and discussion: Our results show that oHSV replication and gene expression results in vitro were not predictive of oHSV direct oncolytic activity in vivo. Tumors that supported viral replication in cell culture studies resisted viral replication by both oHSVs and restricted M002 transgene expression in vivo. Furthermore, two IL-12-oHSVs with equivalent transcriptional activity differed in IL-12 protein production in vivo, and the differences in IL-12 protein levels were reflected in immune infiltrate activity changes as well as tumor growth suppression differences between the IL-12-oHSVs. C002-treated tumors exhibited sustained IL-12 production with improved dendritic cells, monocyte-macrophage activity (MHCII, CD80/CD86 upregulation) and a polyfunctional Th1-cell response in the tumor infiltrates. Conclusion: These results suggest that transgene protein production differences between oHSVs in vivo, in addition to replication differences, can impact OV-therapeutic activity.


Asunto(s)
Interleucina-12 , Viroterapia Oncolítica , Virus Oncolíticos , Transgenes , Replicación Viral , Animales , Interleucina-12/genética , Interleucina-12/metabolismo , Ratones , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Línea Celular Tumoral , Inmunoterapia/métodos , Humanos , Simplexvirus/genética , Células Dendríticas/inmunología , Femenino
5.
J Exp Clin Cancer Res ; 43(1): 193, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992659

RESUMEN

BACKGROUND: Macrophages play important roles in phagocytosing tumor cells. However, tumors escape macrophage phagocytosis in part through the expression of anti-phagocytic signals, most commonly CD47. In Ewing sarcoma (ES), we found that tumor cells utilize dual mechanisms to evade macrophage clearance by simultaneously over-expressing CD47 and down-regulating cell surface calreticulin (csCRT), the pro-phagocytic signal. Here, we investigate the combination of a CD47 blockade (magrolimab, MAG) to inhibit the anti-phagocytic signal and a chemotherapy regimen (doxorubicin, DOX) to enhance the pro-phagocytic signal to induce macrophage phagocytosis of ES cells in vitro and inhibit tumor growth and metastasis in vivo. METHODS: Macrophages were derived from human peripheral blood monocytes by granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF). Flow cytometry- and microscopy-based in-vitro phagocytosis assays were performed to evaluate macrophage phagocytosis of ES cells. Annexin-V assay was performed to evaluate apoptosis. CD47 was knocked out by CRISPR/Cas9 approach. ES cell-based and patient-derived-xenograft (PDX)-based mouse models were utilized to assess the effects of MAG and/or DOX on ES tumor development and animal survival. RNA-Seq combined with CIBERSORTx analysis was utilized to identify changes in tumor cell transcriptome and tumor infiltrating immune cell profiling in MAG and/or DOX treated xenograft tumors. RESULTS: We found that MAG significantly increased macrophage phagocytosis of ES cells in vitro (p < 0.01) and had significant effect on reducing tumor burden (p < 0.01) and increasing survival in NSG mouse model (p < 0.001). The csCRT level on ES cells was significantly enhanced by DOX in a dose- and time-dependent manner (p < 0.01). Importantly, DOX combined with MAG significantly enhanced macrophage phagocytosis of ES cells in vitro (p < 0.01) and significantly decreased tumor burden (p < 0.01) and lung metastasis (p < 0.0001) and extended animal survival in vivo in two different mouse models of ES (p < 0.0001). Furthermore, we identified CD38, CD209, CD163 and CD206 as potential markers for ES-phagocytic macrophages. Moreover, we found increased M2 macrophage infiltration and decreased expression of Cd209 in the tumor microenvironment of MAG and DOX combinatorial therapy treated tumors. CONCLUSIONS: By turning "two keys" simultaneously to reactivate macrophage phagocytic activity, our data demonstrated an effective and highly translatable alternative therapeutic approach utilizing innate (tumor associated macrophages) immunotherapy against high-risk metastatic ES.


Asunto(s)
Inmunoterapia , Macrófagos , Sarcoma de Ewing , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/patología , Sarcoma de Ewing/terapia , Sarcoma de Ewing/tratamiento farmacológico , Animales , Ratones , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Inmunoterapia/métodos , Antígeno CD47/metabolismo , Línea Celular Tumoral , Fagocitosis , Ensayos Antitumor por Modelo de Xenoinjerto , Femenino , Inmunidad Innata , Modelos Animales de Enfermedad
6.
J Virol ; 86(1): 610-4, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22072777

RESUMEN

The chimeric herpes simplex viruses (HSV) are Δγ134.5 vectors encoding the human cytomegalovirus (HCMV) IRS1 or TRS1 genes. They are capable of late viral protein synthesis and are superior to Δγ134.5 HSVs in oncolytic activity. The interferon (IFN) response limits efficient HSV gene expression and replication. HCMV TRS1 and IRS1 restore one γ134.5 gene function: evasion of IFN-inducible protein kinase R, allowing late viral protein synthesis. Here we show that, unlike wild-type HSV, the chimeric HSV do not restore another γ134.5 function, the suppression of early IFN signaling mediated by IFN regulatory factor 3 (IRF3).


Asunto(s)
Citomegalovirus/genética , Herpes Simple/genética , Factor 3 Regulador del Interferón/inmunología , Viroterapia Oncolítica/instrumentación , Simplexvirus/fisiología , Proteínas Virales/genética , Línea Celular , Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vectores Genéticos/fisiología , Herpes Simple/inmunología , Herpes Simple/metabolismo , Herpes Simple/virología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/inmunología , Fosforilación , Simplexvirus/genética , Simplexvirus/inmunología , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Replicación Viral
7.
Mol Ther Oncolytics ; 30: 39-55, 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37583388

RESUMEN

Oncolytic viruses, modified for tumor-restricted infection, are a promising cancer immunotherapeutic, yet much remains to be understood about factors driving their activity and outcome in the tumor microenvironment. Here, we report that oncolytic herpes simplex virus C134, previously found to exert T cell-dependent efficacy in mouse models of glioblastoma, exerts T cell-independent efficacy in mouse models of medulloblastoma, indicating this oncolytic virus uses different mechanisms in different tumors. We investigated C134's behavior in mouse medulloblastomas, using single cell RNA sequencing to map C134-induced gene expression changes across cell types, timepoints, and medulloblastoma subgroup models at whole-transcriptome resolution. Our work details substantial oncolytic virus-induced transcriptional remodeling of medulloblastoma-infiltrating immune cells, 10 subpopulations of monocytes and macrophages collectively demonstrating M1-like responses to C134, and suggests C134 be investigated as a potential new therapy for medulloblastoma.

8.
Pediatr Res ; 71(4 Pt 2): 500-10, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22430386

RESUMEN

Cancer stem cells (CSCs), also termed "cancer-initiating cells" or "cancer progenitor cells," which have the ability to self-renew, proliferate, and maintain the neoplastic clone, have recently been discovered in a wide variety of pediatric tumors. These CSCs are thought to be responsible for tumorigenesis and tumor maintenance, aggressiveness, and recurrence due to inherent resistance to current treatment modalities such as chemotherapy and radiation. Oncolytic virotherapy offers a novel, targeted approach for eradicating pediatric CSCs using mechanisms of cell killing that differ from conventional therapies. Moreover, oncolytic viruses have the ability to target specific features of CSCs such as cell-surface proteins, transcription factors, and the CSC microenvironment. Through genetic engineering, a wide variety of foreign genes may be expressed by oncolytic viruses to augment the oncolytic effect. We review the current data regarding the ability of several types of oncolytic viruses (herpes simplex virus-1, adenovirus, reovirus, Seneca Valley virus, vaccinia virus, Newcastle disease virus, myxoma virus, vesicular stomatitis virus) to target and kill both CSCs and tumor cells in pediatric tumors. We highlight advantages and limitations of each virus and potential ways in which next-generation engineered viruses may target resilient CSCs.


Asunto(s)
Ingeniería Genética/métodos , Neoplasias/prevención & control , Células Madre Neoplásicas/citología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Pediatría/métodos , Niño , Humanos , Células Madre Neoplásicas/virología , Virus Oncolíticos/genética , Pediatría/tendencias
9.
Front Immunol ; 13: 1038096, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36483545

RESUMEN

High grade gliomas are identified as malignant central nervous tumors that spread rapidly and have a universally poor prognosis. Historically high grade gliomas in the pediatric population have been treated similarly to adult high grade gliomas. For the first time, the most recent classification of central nervous system tumors by World Health Organization has divided adult from pediatric type diffuse high grade gliomas, underscoring the biologic differences between these tumors in different age groups. The objective of our review is to compare high grade gliomas in the adult versus pediatric patient populations, highlighting similarities and differences in epidemiology, etiology, pathogenesis and therapeutic approaches. High grade gliomas in adults versus children have varying clinical presentations, molecular biology background, and response to chemotherapy, as well as unique molecular targets. However, increasing evidence show that they both respond to recently developed immunotherapies. This review summarizes the distinctions and commonalities between the two in disease pathogenesis and response to therapeutic interventions with a focus on immunotherapy.


Asunto(s)
Genómica , Neoplasias , Humanos , Niño , Adulto
10.
Clin Cancer Res ; 28(3): 498-506, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35105718

RESUMEN

PURPOSE: Previously, clinical trials of experimental virotherapy for recurrent glioblastoma multiforme (GBM) demonstrated that inoculation with a conditionally replication-competent Δγ134.5 oncolytic herpes simplex virus (oHSV), G207, was safe. Following the initial safety study, a phase Ib trial enrolled 6 adult patients diagnosed with GBM recurrence from which tumor tissue was banked for future studies. PATIENTS AND METHODS: Here, we analyzed tumor RNA sequencing (RNA-seq) data obtained from pre- and posttreatment (collected 2 or 5 days after G207 injection) biopsies from the phase Ib study patients. RESULTS: Using a Spearman rank-order correlation analysis, we identified approximately 500 genes whose expression pattern correlated with survival duration. Many of these genes were enriched for the intrinsic IFN-mediated antiviral and adaptive immune functional responses, including immune cell chemotaxis and antigen presentation to T-cells. Furthermore, we show that the expression of several T-cell-related genes was highest in the patient with the longest survival after G207 inoculation. CONCLUSIONS: Our data support that the oHSV-induced type I IFN production and the subsequent recruitment of an adaptive immune response differed between enrolled patients and showed association with survival duration in patients with recurrent malignant glioma after treatment with an early generation oHSV.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Ensayos Clínicos Fase I como Asunto , Perfilación de la Expresión Génica/métodos , Glioblastoma/genética , Glioblastoma/terapia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos , ARN Neoplásico/genética , Simplexvirus , Adulto , Anciano , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Femenino , Glioblastoma/inmunología , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Tasa de Supervivencia
11.
Mol Pharm ; 8(1): 44-9, 2011 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-21142023

RESUMEN

Conditionally replication-competent Herpes Simplex Virus Type 1 (HSV-1) vectors expressing foreign genes have been developed as experimental therapeutic agents. Traditional methods of virus construction, including growth selection based on thymidine kinase gene expression, and color selection based on a reporter gene expression are often time-consuming and relatively inefficient. This review summarizes the various strategies developed in recent years for the rapid and efficient construction of novel conditionally replication-competent mutant HSV expressing multiple foreign genes. Additionally, two new modifications of existing strategies, which have not been previously reported, are discussed.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Herpesvirus Humano 1/fisiología , Neoplasias/terapia , Replicación Viral/genética , Herpesvirus Humano 1/genética , Humanos , Neoplasias/genética
12.
J Immunother Cancer ; 9(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34599026

RESUMEN

BACKGROUND: Oncolytic virotherapy (OV) is an immunotherapy that incorporates viral cancer cell lysis with engagement of the recruited immune response against cancer cells. Pediatric solid tumors are challenging targets because they contain both an inert immune environment and a quiet antigenic landscape, making them more resistant to conventional OV approaches. Further complicating this, herpes simplex virus suppresses host gene expression during virotherapy infection. METHODS: We therefore developed a multimodal oncolytic herpes simplex virus (oHSV) that expresses ephrin A2 (EphA2), a shared tumor-associated antigen (TAA) expressed by many tumors to improve immune-mediated antitumor activity. We verified the virus genotypically and phenotypically and then tested it in an oHSV-resistant orthotopic model (including immunophenotypic analysis), in flank and in T cell-deficient mouse models. We then assessed the antigen-expressing virus in an unrelated peripheral tumor model that also expresses the shared tumor antigen and evaluated functional T-cell response from the treated mice. RESULTS: Virus-based EphA2 expression induces a robust acquired antitumor immune responses in both an oHSV-resistant murine brain and peripheral tumor model. Our new multimodal oncolytic virus (1) improves survival in viroimmunotherapy resistant tumors, (2) alters both the infiltrating and peripheral T-cell populations capable of suppressing tumor growth on rechallenge, and (3) produces EphA2-specific CD8 effector-like populations. CONCLUSIONS: Our results suggest that this flexible viral-based platform enables immune recognition of the shared TAA and improves the immune-therapeutic response, thus making it well suited for low-mutational load tumors.


Asunto(s)
Herpes Simple/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Animales , Modelos Animales de Enfermedad , Inmunoterapia/métodos , Ratones
13.
Nat Metab ; 2(7): 635-647, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32694789

RESUMEN

T cells undergo metabolic rewiring to meet their bioenergetic, biosynthetic and redox demands following antigen stimulation. To fulfil these needs, effector T cells must adapt to fluctuations in environmental nutrient levels at sites of infection and inflammation. Here, we show that effector T cells can utilize inosine, as an alternative substrate, to support cell growth and function in the absence of glucose in vitro. T cells metabolize inosine into hypoxanthine and phosphorylated ribose by purine nucleoside phosphorylase. We demonstrate that the ribose subunit of inosine can enter into central metabolic pathways to provide ATP and biosynthetic precursors, and that cancer cells display diverse capacities to utilize inosine as a carbon source. Moreover, the supplementation with inosine enhances the anti-tumour efficacy of immune checkpoint blockade and adoptive T-cell transfer in solid tumours that are defective in metabolizing inosine, reflecting the capability of inosine to relieve tumour-imposed metabolic restrictions on T cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Carbono/metabolismo , Glucosa/deficiencia , Inosina/metabolismo , Traslado Adoptivo , Animales , Línea Celular Tumoral , Células HeLa , Humanos , Hipoxantina/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Nutrientes , Purina-Nucleósido Fosforilasa/metabolismo , Ribosa/metabolismo
14.
J Neurooncol ; 95(2): 199-209, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19521665

RESUMEN

Oncolytic herpes simplex viruses (HSV) hold promise for therapy of glioblastoma multiforme (GBM) resistant to traditional therapies. We examined the ability of genetically engineered HSV to infect and kill cells that express CD133, a putative marker of glioma progenitor cells (GPC), to determine if GPC have an inherent therapeutic resistance to HSV. Expression of CD133 and CD111 (nectin-1), the major entry molecule for HSV, was variable in six human glioma xenografts, at initial disaggregation and after tissue culture. Importantly, both CD133+ and CD133- populations of glioma cells expressed CD111 in similar relative proportions in five xenografts, and CD133+ and CD133- glioma cell subpopulations were equally sensitive to killing in vitro by graded dilutions of wild-type HSV-1(F) or several different gamma(1)34.5-deleted viruses. GPC did not display an inherent resistance to HSV. While CD111 expression was an important factor for determining sensitivity of glioma cells to HSV oncolysis, it was not the only factor. Our findings support the notion that HSV will not be able to effectively enter, infect, and kill cells in tumors that have low CD111 expression (<20%). However, virotherapy with HSV may be very effective against CD111+ GPC resistant to traditional therapies.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Encefálicas/terapia , Moléculas de Adhesión Celular/metabolismo , Glioblastoma/terapia , Glicoproteínas/metabolismo , Herpesvirus Humano 1/fisiología , Viroterapia Oncolítica , Péptidos/metabolismo , Antígeno AC133 , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Efecto Citopatogénico Viral , Ingeniería Genética/métodos , Vectores Genéticos , Glioblastoma/metabolismo , Glioblastoma/virología , Humanos , Ratones , Ratones Desnudos , Nectinas , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Ther Oncolytics ; 15: 91-100, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31650029

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are an aggressive soft-tissue sarcoma amenable only to surgical resection. Oncolytic herpes simplex viruses (oHSVs) are a promising experimental therapy. We previously showed that basal interferon (IFN) and nuclear factor κB (NFκB) signaling upregulate IFN-stimulated gene (ISG) expression and restrict efficient viral infection and cell-to-cell spread in ∼50% of tested MPNSTs. Stimulator of Interferon Genes (STING) integrates DNA sensor activity and mediates downstream IFN signaling in infected cells. We sought to identify STING's role in oHSV resistance and contribution to basal ISG upregulation in MPNSTs. We show that the level of STING activity in human MPNST cell lines is predictive of oHSV sensitivity and that resistant cell lines have intact mechanisms for detection of cytosolic double-stranded DNA (dsDNA). Furthermore, we show that STING downregulation renders MPNSTs more permissive to oHSV infection and cell-to-cell spread. While next-generation viruses can exploit this loss of STING activity, first-generation viruses remain restricted. Finally, STING is not integral to the previously-observed basal ISG upregulation, indicating that other pathways contribute to basal IFN signaling in resistant MPNSTs. These data broaden our understanding of the intrinsic pathways in MPNSTs and their role in oHSV resistance and offer potential targets to potentiate oncolytic virus activity.

16.
Mol Ther Oncolytics ; 13: 7-13, 2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-30989105

RESUMEN

A network of pattern recognition receptors (PRRs) is responsible for the detection of invading viruses and acts as the trigger for the host antiviral response. Central to this apparatus is stimulator of interferon genes (STING), which functions as a node and integrator of detection signals. Owing to its role in both intrinsic and adaptive immunity, STING has become a focus for researchers in the field of oncolytic virotherapy. In this review, we consider the function of the cGAS-STING axis and its regulation, both by cellular mechanisms and as a result of viral interference.

17.
Mol Ther Oncolytics ; 13: 14-21, 2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-30997392

RESUMEN

Oncolytic viral therapy has gained significant traction as cancer therapy over the past 2 decades. Oncolytic viruses are uniquely designed both to lyse tumor cells through their replication and to recruit immune responses against virally infected cells. Increasingly, investigators are leveraging this immune response to target the immunosuppressive tumor microenvironment and improve immune effector response against bystander tumor cells. In this article, we review the spectrum of preclinical, early-stage clinical, and potential future efforts with cytokine-secreting oncolytic viruses, with a focus on the treatment of brain tumors and solid tumors.

18.
Mol Ther Oncolytics ; 15: 38-48, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31650024

RESUMEN

Solid tumors contain a mixture of malignant cells and non-malignant infiltrating cells that often create a chronic inflammatory and immunosuppressive microenvironment that restricts immunotherapeutic approaches. Although childhood and adult cancers share some similarities related to microenvironmental changes, pediatric cancers are unique, and adult cancer practices may not be wholly applicable to our pediatric patients. This review highlights the differences in tumorigenesis, viral infection, and immunologic response between children and adults that need to be considered when trying to apply experiences from experimental therapies in adult cancer patients to pediatric cancers.

19.
Pediatr Infect Dis J ; 27(10): 946-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18776818

RESUMEN

A 21-month-old girl with neuroblastoma developed chronic verrucous Oka strain varicella-zoster infection during chemotherapy. Virus isolated from the patient demonstrated high-level acyclovir resistance, and its thymidine kinase had no in vitro enzymatic activity. After foscarnet therapy, she underwent stem cell transplantation without varicella reactivation. This is only the second reported case of resistant varicella zoster virus caused by Oka strain virus.


Asunto(s)
Aciclovir/uso terapéutico , Vacuna contra la Varicela/efectos adversos , Varicela/etiología , Herpesvirus Humano 3/efectos de los fármacos , Neuroblastoma/complicaciones , Antineoplásicos/uso terapéutico , Antivirales/uso terapéutico , Varicela/tratamiento farmacológico , Enfermedad Crónica , Farmacorresistencia Viral , Femenino , Humanos , Huésped Inmunocomprometido , Lactante , Neuroblastoma/tratamiento farmacológico
20.
Cancer Immunol Res ; 6(12): 1499-1510, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30352799

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas resistant to most cancer treatments. Surgical resection remains the primary treatment, but this is often incomplete, ultimately resulting in high mortality and morbidity rates. There has been a resurgence of interest in oncolytic virotherapy because of encouraging preclinical and clinical trial results. Oncolytic herpes simplex virus (oHSV) selectively replicates in cancer cells, lysing the cell and inducing antitumor immunity. We previously showed that basal interferon (IFN) signaling increases interferon-stimulated gene (ISG) expression, restricting viral replication in almost 50% of MPNSTs. The FDA-approved drug ruxolitinib (RUX) temporarily resets this constitutively active STAT signaling and renders the tumor cells susceptible to oHSV infection in cell culture. In the studies described here, we translated our in vitro results into a syngeneic MPNST tumor model. Consistent with our previous results, murine MPNSTs exhibit a similar IFN- and ISG-mediated oHSV-resistance mechanism, and virotherapy alone provides no antitumor benefit in vivo However, pretreatment of mice with ruxolitinib reduced ISG expression, making the tumors susceptible to oHSV infection. Ruxolitinib pretreatment improved viral replication and altered the oHSV-induced immune-mediated response. Our results showed that this combination therapy increased CD8+ T-cell activation in the tumor microenvironment and that this population was indispensable for the antitumor benefit that follows from the combination of RUX and oHSV. These data suggest that JAK inhibition prior to oncolytic virus treatment augments both oHSV replication and the immunotherapeutic efficacy of oncolytic herpes virotherapy.


Asunto(s)
Herpesvirus Humano 1/efectos de los fármacos , Neoplasias de la Vaina del Nervio/terapia , Viroterapia Oncolítica/métodos , Pirazoles/farmacología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Terapia Combinada , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Interferones/metabolismo , Interferones/farmacología , Ratones Endogámicos C57BL , Neoplasias de la Vaina del Nervio/inmunología , Neoplasias de la Vaina del Nervio/mortalidad , Nitrilos , Virus Oncolíticos/efectos de los fármacos , Virus Oncolíticos/fisiología , Pirimidinas , Tasa de Supervivencia , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA