Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Methods Mol Biol ; 2423: 165-177, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34978698

RESUMO

Oncolytic virotherapy translational research in the current era is heavily focused on the interaction of the immune system and tumor microenvironment with oncolytic viruses. Preclinical xenograft studies using human cells in immunodeficient mouse models does not serve this purpose. As a consequence, developing syngeneic immunocompetent murine cancer models sensitive to infection and growth of specific oncolytic viruses is required. The group 3 subtype of medulloblastoma, among the four molecular subgroups-WNT, SHH, Group 3, and Group 4, has the worst prognosis and the poorest outcome. Sadly, current treatments cause long-term toxicity and morbidity to survivors adversely affecting their quality of life. Alternate effective therapy with less side effects is urgently needed. We have shown that oncolytic measles virus (MV) is effective against localized as well as CSF-disseminated medulloblastoma in immunodeficient mouse models. To study the interaction of immune system with oncolytic measles virotherapy, we have developed a murine group 3 medulloblastoma cell line (CSCG) that is infectible by MV, is killed by MV, allows replication of MV, and is tumorigenic in the brain of syngeneic transgenic immune-competent mice. Intratumoral injection of MV results in significant prolongation of survival in mice bearing CSCG tumors in the brain. This model provides the first suitable platform to examine therapeutic regimens of MV therapy for MB tumors in the presence of intact immune system. Here, we describe our lab protocols to develop this cell line and the mouse model.


Assuntos
Neoplasias Cerebelares , Sarampo , Meduloblastoma , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Linhagem Celular Tumoral , Neoplasias Cerebelares/terapia , Humanos , Sarampo/terapia , Vírus do Sarampo/genética , Meduloblastoma/patologia , Meduloblastoma/terapia , Camundongos , Camundongos Nus , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Qualidade de Vida , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Brain Pathol ; 31(4): e12918, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33141488

RESUMO

Intracranial mesenchymal tumors with FET-CREB fusions are a recently described group of neoplasms in children and young adults characterized by fusion of a FET family gene (usually EWSR1, but rarely FUS) to a CREB family transcription factor (ATF1, CREB1, or CREM), and have been variously termed intracranial angiomatoid fibrous histiocytoma or intracranial myxoid mesenchymal tumor. The clinical outcomes, histologic features, and genomic landscape are not well defined. Here, we studied 20 patients with intracranial mesenchymal tumors proven to harbor FET-CREB fusion by next-generation sequencing (NGS). The 16 female and four male patients had a median age of 14 years (range 4-70). Tumors were uniformly extra-axial or intraventricular and located at the cerebral convexities (n = 7), falx (2), lateral ventricles (4), tentorium (2), cerebellopontine angle (4), and spinal cord (1). NGS demonstrated that eight tumors harbored EWSR1-ATF1 fusion, seven had EWSR1-CREB1, four had EWSR1-CREM, and one had FUS-CREM. Tumors were uniformly well circumscribed and typically contrast enhancing with solid and cystic growth. Tumors with EWSR1-CREB1 fusions more often featured stellate/spindle cell morphology, mucin-rich stroma, and hemangioma-like vasculature compared to tumors with EWSR1-ATF1 fusions that most often featured sheets of epithelioid cells with mucin-poor collagenous stroma. These tumors demonstrated polyphenotypic immunoprofiles with frequent positivity for desmin, EMA, CD99, MUC4, and synaptophysin, but absence of SSTR2A, myogenin, and HMB45 expression. There was a propensity for local recurrence with a median progression-free survival of 12 months and a median overall survival of greater than 60 months, with three patients succumbing to disease (all with EWSR1-ATF1 fusions). In combination with prior case series, this study provides further insight into intracranial mesenchymal tumors with FET-CREB fusion, which represent a distinct group of CNS tumors encompassing both intracranial myxoid mesenchymal tumor and angiomatoid fibrous histiocytoma-like neoplasms.


Assuntos
Neoplasias Encefálicas/patologia , Histiocitoma Fibroso Benigno/patologia , Histiocitoma Fibroso Maligno/patologia , Proteínas de Fusão Oncogênica/metabolismo , Adolescente , Adulto , Idoso , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Feminino , Fusão Gênica/genética , Histiocitoma Fibroso Benigno/diagnóstico , Histiocitoma Fibroso Benigno/metabolismo , Histiocitoma Fibroso Maligno/diagnóstico , Histiocitoma Fibroso Maligno/genética , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica/genética , Adulto Jovem
3.
Acta Neuropathol Commun ; 8(1): 151, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859279

RESUMO

The FGFR1 gene encoding fibroblast growth factor receptor 1 has emerged as a frequently altered oncogene in the pathogenesis of multiple low-grade neuroepithelial tumor (LGNET) subtypes including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumor (DNT), rosette-forming glioneuronal tumor (RGNT), and extraventricular neurocytoma (EVN). These activating FGFR1 alterations in LGNET can include tandem duplication of the exons encoding the intracellular tyrosine kinase domain, in-frame gene fusions most often with TACC1 as the partner, or hotspot missense mutations within the tyrosine kinase domain (either at p.N546 or p.K656). However, the specificity of these different FGFR1 events for the various LGNET subtypes and accompanying genetic alterations are not well defined. Here we performed comprehensive genomic and epigenomic characterization on a diverse cohort of 30 LGNET with FGFR1 alterations. We identified that RGNT harbors a distinct epigenetic signature compared to other LGNET with FGFR1 alterations, and is uniquely characterized by FGFR1 kinase domain hotspot missense mutations in combination with either PIK3CA or PIK3R1 mutation, often with accompanying NF1 or PTPN11 mutation. In contrast, EVN harbors its own distinct epigenetic signature and is characterized by FGFR1-TACC1 fusion as the solitary pathogenic alteration. Additionally, DNT and pilocytic astrocytoma are characterized by either kinase domain tandem duplication or hotspot missense mutations, occasionally with accompanying NF1 or PTPN11 mutation, but lacking the accompanying PIK3CA or PIK3R1 mutation that characterizes RGNT. The glial component of LGNET with FGFR1 alterations typically has a predominantly oligodendroglial morphology, and many of the pilocytic astrocytomas with FGFR1 alterations lack the biphasic pattern, piloid processes, and Rosenthal fibers that characterize pilocytic astrocytomas with BRAF mutation or fusion. Together, this analysis improves the classification and histopathologic stratification of LGNET with FGFR1 alterations.


Assuntos
Neoplasias Neuroepiteliomatosas/classificação , Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Neoplasias da Medula Espinal/classificação , Neoplasias da Medula Espinal/genética , Neoplasias da Medula Espinal/patologia , Adulto Jovem
4.
Acta Neuropathol ; 139(6): 1071-1088, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32303840

RESUMO

Brain tumors are the most common solid tumors of childhood, and the genetic drivers and optimal therapeutic strategies for many of the different subtypes remain unknown. Here, we identify that bithalamic gliomas harbor frequent mutations in the EGFR oncogene, only rare histone H3 mutation (in contrast to their unilateral counterparts), and a distinct genome-wide DNA methylation profile compared to all other glioma subtypes studied to date. These EGFR mutations are either small in-frame insertions within exon 20 (intracellular tyrosine kinase domain) or missense mutations within exon 7 (extracellular ligand-binding domain) that occur in the absence of accompanying gene amplification. We find these EGFR mutations are oncogenic in primary astrocyte models and confer sensitivity to specific tyrosine kinase inhibitors dependent on location within the kinase domain or extracellular domain. We initiated treatment with targeted kinase inhibitors in four children whose tumors harbor EGFR mutations with encouraging results. This study identifies a promising genomically-tailored therapeutic strategy for bithalamic gliomas, a lethal and genetically distinct brain tumor of childhood.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Glioma/genética , Mutação/genética , Adolescente , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Epigênese Genética/genética , Receptores ErbB/genética , Feminino , Glioma/tratamento farmacológico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Inibidores de Proteínas Quinases/farmacologia
6.
Brain Pathol ; 30(1): 46-62, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31104347

RESUMO

High-grade neuroepithelial tumor with BCOR exon 15 internal tandem duplication (HGNET BCOR ex15 ITD) is a recently proposed tumor entity of the central nervous system (CNS) with a distinct methylation profile and characteristic genetic alteration. The complete spectrum of histologic features, accompanying genetic alterations, clinical outcomes, and optimal treatment for this new tumor entity are largely unknown. Here, we performed a comprehensive assessment of 10 new cases of HGNET BCOR ex15 ITD. The tumors mostly occurred in young children and were located in the cerebral or cerebellar hemispheres. On imaging all tumors were large, well-circumscribed, heterogeneous masses with variable enhancement and reduced diffusion. They were histologically characterized by predominantly solid growth, glioma-like fibrillarity, perivascular pseudorosettes, and palisading necrosis, but absence of microvascular proliferation. They demonstrated sparse to absent GFAP expression, no synaptophysin expression, variable OLIG2 and NeuN positivity, and diffuse strong BCOR nuclear positivity. While BCOR exon 15 internal tandem duplication was the solitary pathogenic alteration identified in six cases, four cases contained additional alterations including CDKN2A/B homozygous deletion, TERT amplification or promoter hotspot mutation, and damaging mutations in TP53, BCORL1, EP300, SMARCA2 and STAG2. While the limited clinical follow-up in prior reports had indicated a uniformly dismal prognosis for this tumor entity, this cohort includes multiple long-term survivors. Our study further supports inclusion of HGNET BCOR ex15 ITD as a distinct CNS tumor entity and expands the known clinicopathologic, radiographic, and genetic features.


Assuntos
Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Adolescente , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Neoplasias do Sistema Nervoso Central/genética , Criança , Pré-Escolar , Inibidor p16 de Quinase Dependente de Ciclina/genética , Proteína p300 Associada a E1A/genética , Éxons , Feminino , Genômica , Glioma/genética , Humanos , Lactente , Estimativa de Kaplan-Meier , Masculino , Fator de Transcrição 2 de Oligodendrócitos/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Telomerase/genética , Fatores de Transcrição/genética
8.
Mayo Clin Proc ; 94(9): 1834-1839, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31235278

RESUMO

Recent measles epidemics in US and European cities where vaccination coverage has declined are providing a harsh reminder for the need to maintain protective levels of immunity across the entire population. Vaccine uptake rates have been declining in large part because of public misinformation regarding a possible association between measles vaccination and autism for which there is no scientific basis. The purpose of this article is to address a new misinformed antivaccination argument-that measles immunity is undesirable because measles virus is protective against cancer. Having worked for many years to develop engineered measles viruses as anticancer therapies, we have concluded (1) that measles is not protective against cancer and (2) that its potential utility as a cancer therapy will be enhanced, not diminished, by prior vaccination.


Assuntos
Comunicação , Vírus do Sarampo/imunologia , Sarampo/epidemiologia , Sarampo/prevenção & controle , Terapia Viral Oncolítica/métodos , Vacinação/efeitos adversos , Criança , Pré-Escolar , Controle de Doenças Transmissíveis/organização & administração , Europa (Continente) , Feminino , Humanos , Masculino , Prevalência , Medição de Risco , Estados Unidos , Vacinação/métodos
10.
Acta Neuropathol ; 137(1): 139-150, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30196423

RESUMO

Radiotherapy improves survival for common childhood cancers such as medulloblastoma, leukemia, and germ cell tumors. Unfortunately, long-term survivors suffer sequelae that can include secondary neoplasia. Gliomas are common secondary neoplasms after cranial or craniospinal radiation, most often manifesting as high-grade astrocytomas with poor clinical outcomes. Here, we performed genetic profiling on a cohort of 12 gliomas arising after therapeutic radiation to determine their molecular pathogenesis and assess for differences in genomic signature compared to their spontaneous counterparts. We identified a high frequency of TP53 mutations, CDK4 amplification or CDKN2A homozygous deletion, and amplifications or rearrangements involving receptor tyrosine kinase and Ras-Raf-MAP kinase pathway genes including PDGFRA, MET, BRAF, and RRAS2. Notably, all tumors lacked alterations in IDH1, IDH2, H3F3A, HIST1H3B, HIST1H3C, TERT (including promoter region), and PTEN, which genetically define the major subtypes of diffuse gliomas in children and adults. All gliomas in this cohort had very low somatic mutation burden (less than three somatic single nucleotide variants or small indels per Mb). The ten high-grade gliomas demonstrated markedly aneuploid genomes, with significantly increased quantity of intrachromosomal copy number breakpoints and focal amplifications/homozygous deletions compared to spontaneous high-grade gliomas, likely as a result of DNA double-strand breaks induced by gamma radiation. Together, these findings demonstrate a distinct molecular pathogenesis of secondary gliomas arising after radiation therapy and identify a genomic signature that may aid in differentiating these tumors from their spontaneous counterparts.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Glioma/genética , Glioma/radioterapia , Adolescente , Adulto , Astrocitoma/radioterapia , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/radioterapia , Criança , Pré-Escolar , Feminino , Genômica , Homozigoto , Humanos , Masculino , Mutação/genética , Deleção de Sequência/genética , Telomerase/genética , Adulto Jovem
12.
Neuro Oncol ; 20(12): 1606-1615, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-29912438

RESUMO

Background: Oncolytic measles virus (MV) is effective in xenograft models of many tumor types in immune-compromised mice. However, no murine cell line exists that is tumorigenic, grows in immune-competent mice, and is killed by MV. The lack of such a model prevents an examination of the effect of the immune system on MV oncotherapy. Methods: Cerebellar stem cells from human CD46-transgenic immunocompetent mice were transduced to express Sendai virus C-protein, murine C-Myc, and Gfi1b proteins. The resultant cells were injected into the brain of NSG mice, and a cell line, called CSCG, was prepared from the resulting tumor. Results: CSCG cells are highly proliferative, and express stem cell markers. These cells are permissive for replication of MV and are killed by the virus in a dose- and time-dependent manner. CSCG cells form aggressive tumors that morphologically resemble medulloblastoma when injected into the brains of immune-competent mice. On the molecular level, CSCG tumors overexpress natriuretic peptide receptor 3 and gamma-aminobutyric acid type A receptor alpha 5, markers of Group 3 medulloblastoma. A single intratumoral injection of MV‒green fluorescent protein resulted in complete tumor regression and prolonged survival of animals compared with treatments with phosphate buffered saline (P = 0.0018) or heat-inactivated MV (P = 0.0027). Conclusions: This immune-competent model provides the first platform to test therapeutic regimens of oncolytic MV for Group 3 medulloblastoma in the presence of anti-measles immunity. The strategy presented here can be used to make MV-sensitive murine models of any human tumor for which the driving mutations are known.


Assuntos
Neoplasias Cerebelares/terapia , Modelos Animais de Doenças , Imunocompetência , Vírus do Sarampo/genética , Meduloblastoma/terapia , Terapia Viral Oncolítica , Animais , Neoplasias Cerebelares/imunologia , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/virologia , Humanos , Sarampo/virologia , Meduloblastoma/imunologia , Meduloblastoma/metabolismo , Meduloblastoma/virologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Células Tumorais Cultivadas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Neuro Oncol ; 19(5): 699-709, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28453743

RESUMO

Background: Molecular profiling is revolutionizing cancer diagnostics and leading to personalized therapeutic approaches. Herein we describe our clinical experience performing targeted sequencing for 31 pediatric neuro-oncology patients. Methods: We sequenced 510 cancer-associated genes from tumor and peripheral blood to identify germline and somatic mutations, structural variants, and copy number changes. Results: Genomic profiling was performed on 31 patients with tumors including 11 high-grade gliomas, 8 medulloblastomas, 6 low-grade gliomas, 1 embryonal tumor with multilayered rosettes, 1 pineoblastoma, 1 uveal ganglioneuroma, 1 choroid plexus carcinoma, 1 chordoma, and 1 high-grade neuroepithelial tumor. In 25 cases (81%), results impacted patient management by: (i) clarifying diagnosis, (ii) identifying pathogenic germline mutations, or (iii) detecting potentially targetable alterations. The pathologic diagnosis was amended after genomic profiling for 6 patients (19%), including a high-grade glioma to pilocytic astrocytoma, medulloblastoma to pineoblastoma, ependymoma to high-grade glioma, and medulloblastoma to CNS high-grade neuroepithelial tumor with BCOR alteration. Multiple patients had pathogenic germline mutations, many of which were previously unsuspected. Potentially targetable alterations were identified in 19 patients (61%). Additionally, novel likely pathogenic alterations were identified in 3 cases: an in-frame RAF1 fusion in a BRAF wild-type pleomorphic xanthoastrocytoma, an inactivating ASXL1 mutation in a histone H3 wild-type diffuse pontine glioma, and an in-frame deletion within exon 2 of MAP2K1 in a low-grade astrocytic neoplasm. Conclusions: Our experience demonstrates the significant impact of molecular profiling on diagnosis and treatment of pediatric brain tumors and confirms its feasibility for use at the time of diagnosis or recurrence.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamento farmacológico , Mutação em Linhagem Germinativa , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Terapia de Alvo Molecular , Adolescente , Adulto , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Prognóstico , Adulto Jovem
15.
Neurooncol Pract ; 4(3): 138-150, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29692919

RESUMO

Medulloblastoma is the most common malignant brain tumor affecting children. These tumors are high grade with propensity to metastasize within the central nervous system and, less frequently, outside the neuraxis. Recent advancements in molecular subgrouping of medulloblastoma refine diagnosis and improve counseling in regards to overall prognosis. Both are predicated on the molecular drivers of each subgroup-WNT-activated, SHH-activated, group 3, and group 4. The traditional therapeutic mainstay for medulloblastoma includes a multimodal approach with surgery, radiation, and multiagent chemotherapy. As we discover more about the molecular basis of medulloblastoma, efforts to adjust treatment approaches based on molecular risk stratification are under active investigation. Certainly, the known neurological, developmental, endocrine, and psychosocial injury related to medulloblastoma and its associated therapies motivate ongoing research towards improving treatment for this life-threatening tumor while at the same time minimizing long-term side effects.

16.
Cureus ; 9(12): e1954, 2017 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-29487768

RESUMO

Slipped capital femoral epiphysis (SCFE) is a fracture that results from displacement of the proximal femoral epiphysis from the femoral neck. SCFE can be caused by various endocrinopathies that lead to bone weakening in both adult and pediatric patients. We report a rare case of suprasellar germinoma presenting with SCFE in an 11-year-old female patient. The findings of this case further support the need to consider pituitary lesions as the underlying cause of endocrine deficiences leading to SCFE.

17.
Mol Ther Oncolytics ; 7: 57-66, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29367943

RESUMO

Modified measles virus (MV) has effective oncolytic activity preclinically and is currently being investigated in clinical trials for various types of cancer. We investigated the use of cystine knot proteins (CKPs) to direct MV activity. CKPs are short polypeptides that bind their targets with high affinity. We used a CKP that binds αvß3, αvß5, and α5ß1 integrins with single-digit nanomolar affinity to retarget MV to the integrins (MV-CKPint). MV-CKPint infected, replicated in, and killed human glioblastoma, medulloblastoma, diffuse intrinsic pontine glioma (DIPG), and melanoma cancer cells in vitro, all of which express the target integrins. MV-CKPint activity was competitively blocked by echistatin, an integrin binding peptide. When the CKP was cleaved from the viral H protein at an included protease site, virus activity was abrogated. When delivered intravenously (i.v.), the retargeted virus reached a subcutaneous glioblastoma tumor bed and produced cytopathic effects similar to that shown by intratumoral injection of the virus. Because these target integrins are overexpressed by tumor vascular endothelium, MV-CKPint may allow for effective therapy with i.v. injection. These results indicate for the first time that CKPs can be used to retarget MV for a receptor of choice. In addition, MV-CKPint provides proof of principle for the use of a CKP of interest to retarget any enveloped virus for both oncolytic and gene therapy purposes.

18.
Hum Gene Ther Clin Dev ; 27(4): 145-151, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27604429

RESUMO

The modified Edmonston vaccine strain of measles virus (MV) has shown potent oncolytic efficacy against various tumor types and is being investigated in clinical trials. Our laboratory showed that MV effectively kills medulloblastoma tumor cells in both localized disease and when tumor cells are disseminated through cerebrospinal fluid (CSF). Although the safety of repeated intracerebral injection of modified MV in rhesus macaques has been established, the safety of administering MV into CSF has not been adequately investigated. In this study, we assessed the safety of MV-NIS (MV modified to express the human sodium iodide symporter protein) injected into the CSF of measles-immunized and measles virus-susceptible transgenic (CD46, IFNαRko) mice. Treated animals were administered a single intraventricular injection of 1 × 105 or 1 × 106 TCID50 (50% tissue culture infective dose) of MV-NIS. Detailed clinical observation was performed over a 90-day period. Clinically, we did not observe any measles-related toxic effects or behavioral abnormality in animals of any treated cohort. The complete blood count and blood chemistry analysis results were found to be within normal range for all the cohorts. Histologic examination of brains and spinal cords revealed inflammatory changes, mostly related to the needle track; these resolved by day 21 postinjection. To assess viral biodistribution, quantitative RT-PCR to detect the measles virus N-protein was performed on blood and brain samples. Viral RNA was not detectable in the blood as soon as 2 days after injection, and virus cleared from the brain by 45 days postadministration in all treatment cohorts. In conclusion, our data suggest that a single injection of modified MV into the CSF is safe and can be used in future therapeutic applications.


Assuntos
Vírus do Sarampo/patogenicidade , Sarampo/terapia , Proteína Cofatora de Membrana/fisiologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Receptor de Interferon alfa e beta/fisiologia , Simportadores/fisiologia , Animais , Feminino , Humanos , Injeções Intraventriculares , Masculino , Sarampo/imunologia , Sarampo/virologia , Camundongos , Camundongos Transgênicos
19.
Neuro Oncol ; 17(12): 1568-77, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25838138

RESUMO

BACKGROUND: Atypical teratoid rhabdoid tumor (AT/RT) is a rare, highly malignant pediatric tumor of the central nervous system that is usually refractory to available treatments. The aggressive growth, propensity to disseminate along the neuroaxis, and young age at diagnosis contribute to the poor prognosis. Previous studies have demonstrated the efficacy of using oncolytic measles virus (MV) against localized and disseminated models of medulloblastoma. The purpose of this study was to evaluate the oncolytic potential of MV in experimental models of AT/RT. METHODS: Following confirmation of susceptibility to MV infection and killing of AT/RT cells in vitro, nude mice were injected with BT-12 and BT-16 AT/RT cells stereotactically into the caudate nucleus (primary tumor model) or lateral ventricle (disseminated tumor model). Recombinant MV was administered either intratumorally or intravenously. Survival was determined for treated and control animals. Necropsy was performed on animals showing signs of progressive disease. RESULTS: All cell lines exhibited significant killing when infected with MV, all formed syncytia with infection, and all generated infectious virus after infection. Orthotopic xenografts displayed cells with rhabdoid-like cellular morphology, were negative for INI1 expression, and showed dissemination within the intracranial and spinal subarachnoid spaces. Intratumoral injection of live MV significantly prolonged the survival of animals with intracranial and metastatic tumors. CONCLUSION: These data demonstrate that AT/RT is susceptible to MV killing and suggest that the virus may have a role in treating this tumor in the clinical setting.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Tumor Rabdoide/virologia , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Tumor Rabdoide/terapia , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Oncolytic Virother ; 4: 109-18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27512675

RESUMO

A successful oncolytic virus is one that selectively propagates and destroys cancerous tissue without causing excessive damage to the normal surrounding tissue. Oncolytic measles virus (MV) is one such virus that exhibits this characteristic and thus has rapidly emerged as a potentially useful anticancer modality. Derivatives of the Edmonston MV vaccine strain possess a remarkable safety record in humans. Promising results in preclinical animal models and evidence of biological activity in early phase trials contribute to the enthusiasm. Genetic modifications have enabled MV to evolve from a vaccine agent to a potential anticancer therapy. Specifically, alterations of the MV genome have led to improved tumor selectivity and delivery, therapeutic potency, and immune system modulation. In this article, we will review the advancements that have been made in the design and development of MV that have led to its use as a cancer therapy. In addition, we will discuss the evidence supporting its use, as well as the challenges associated with MV as a potential cancer therapeutic.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA