Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hum Vaccin Immunother ; 19(2): 2262639, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37786375

RESUMO

Schwannomas are slow-growing benign peripheral nerve sheath tumors derived from Schwann-lineage cells that develop in association with NF2-related schwannomatosis (NF2) and schwannomatosis (NF3), as well as spontaneously. Individuals affected with NF2 and NF3 have multiple schwannomas with tumors arising throughout life. Surgical resection, the standard management, is limited in scope and efficacy and is itself associated with significant morbidity. We have previously shown that direct intratumoral injection of attenuated Salmonella Typhimurium (S. Typhimurium), strain VNP20009, showed a potent anti-tumor effect in preclinical NF-2 schwannoma models. The United States Federal Drug Administration (FDA) requires that bacterial products utilized in clinical trials be produced without exposure to animal-derived-products. In this context, we developed, characterized, and tested the antitumor efficacy of an attenuated S. Typhimurium serially passaged in animal-product-free media, naming it VNP20009-AF for "VNP20009-animal-product-free." Our in vitro data did not indicate any significant changes in the viability, motility, or morphology of VNP20009-AF, compared to its parental strain. In vivo efficacy data demonstrated that VNP20009-AF and VNP20009 controlled tumor growth to the same degree in both human NF2-schwannoma xenograft and murine-NF2 schwannoma allograft models. Together, these data support the use of VNP20009-AF for the translation of bacterial schwannoma therapy into clinical trials.


Assuntos
Neurilemoma , Neoplasias Cutâneas , Humanos , Camundongos , Animais , Salmonella typhimurium , Neurilemoma/terapia
2.
Anticancer Res ; 43(1): 1-6, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36585175

RESUMO

BACKGROUND/AIM: This study evaluated the effect of enrofloxacin antibiotic treatment on the ability of an attenuated Salmonella typhimurium (S. typhimurium) strain VNP20009 to control schwannoma growth in a preclinical mouse schwannoma tumor model. MATERIALS AND METHODS: The antitumor efficacy of VNP20009 intratumoral (i.t.) injection was assessed in a syngeneic mouse-NF2 schwannoma model, with and without subcutaneous (s.c.) injection of enrofloxacin beginning on day-1 or day-8 post-VNP20009 injection. S. typhimurium colonization was assessed in excised tumor samples following antibiotic treatment. RESULTS: I.t. injection of the VNP20009 significantly decreased the growth of schwannoma tumors in mice compared to PBS-treated controls. Treatment of mice with enrofloxacin on day-1 post-VNP20009 injection resulted in abrogation of VNP20009-mediated tumor growth control. In contrast, tumor growth in i.t. VNP20009-injected mice infused with enrofloxacin beginning on day 8 was significantly decreased compared to i.t. PBS-injected controls. Enrofloxacin significantly reduced the number of viable VNP20009 bacteria in excised tumor samples within one day of antibiotic infusion. Viable bacteria were either few or essentially eliminated at the end of the experiment in antibiotic-treated animals compared to VNP20009-only. CONCLUSION: Viable VNP20009 can persist for as long as 2.5 weeks following intratumoral injection of schwannoma, during which time tumor growth is retarded. Antibiotic treatment starting 1-day following i.t. VNP20009 abrogated bacterial tumor growth control, whereas initiation of antibiotics 8-days following i.t. VNP20009 was associated with control of tumor growth, albeit less than seen in animals unexposed to antibiotics.


Assuntos
Neurilemoma , Salmonella typhimurium , Camundongos , Animais , Enrofloxacina , Neurilemoma/tratamento farmacológico , Antibacterianos/farmacologia
3.
Proc Natl Acad Sci U S A ; 119(24): e2202719119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35675425

RESUMO

Schwannomas are slow-growing benign neoplasms that develop throughout the body causing pain, sensory/motor dysfunction, and death. Because bacterial immunotherapy has been used in the treatment of some malignant neoplasms, we evaluated attenuated Salmonella typhimurium strains as immunotherapies for benign murine schwannomas. Several bacterial strains were tested, including VNP20009, a highly attenuated strain that was previously shown to be safe in human subjects with advanced malignant neoplasms, and a VNP20009 mutant that was altered in motility and other properties that included adherence and invasion of cultured mammalian cells. VNP20009 controlled tumor growth in two murine schwannoma models and induced changes in cytokine and immune effector cell profiles that were consistent with induction of enhanced innate and adaptive host immune responses compared with controls. Intratumoral (i.t.) injection of S. typhimurium led to tumor cell apoptosis, decreased tumor angiogenesis, and lower growth of the injected schwannoma tumors. Invasive VNP20009 was significantly more efficacious than was a noninvasive derivative in controlling the growth of injected tumors. Bacterial treatment apparently induced systemic antitumor immunity in that the growth of rechallenge schwannomas implanted following primary bacterial treatment was also reduced. Checkpoint programmed death-1 (PD-1) blockade induced by systemic administration of anti-PD-1 antibodies controlled tumor growth to the same degree as i.t. injection of S. typhimurium, and together, these two therapies had an additive effect on suppressing schwannoma growth. These experiments represent validation of a bacterial therapy for a benign neoplasm and support development of S. typhimurium VNP20009, potentially in combination with PD-1 inhibition, as a schwannoma immunotherapy.


Assuntos
Imunoterapia , Neurilemoma , Salmonella typhimurium , Animais , Apoptose , Humanos , Imunoterapia/métodos , Injeções Intralesionais , Camundongos , Neoplasias Experimentais/terapia , Neurilemoma/terapia , Receptor de Morte Celular Programada 1 , Salmonella typhimurium/genética
4.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35055004

RESUMO

Schwannomas are tumors derived from Schwann-lineage cells, cells that protect and support myelinated nerves in the peripheral nervous system. They are typically slow-growing, encapsulated and benign. These tumors develop along peripheral, spinal and cranial nerves causing pain, sensory-motor dysfunction and death. Primary treatment for schwannoma is operative resection which can be associated with significant morbidity. Pharmacotherapy is largely restricted to bevacizumab, which has minimal or no efficacy for many patients and can be associated with treatment-limiting adverse effects. Given the suffering and morbidity associated with schwannoma and the paucity of therapeutic options, there is an urgent need for safe and effective therapies for schwannomas. We previously demonstrated that adeno-associated virus serotype 1 (AAV1) vector mediated delivery of the inflammasome adaptor protein, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) under the control of the P0 promoter, produced a prolonged reduction in tumor volume and tumor-associated pain in human xenograft and mouse syngeneic schwannoma models. Here, we present data essential for the translation of our AAV1-P0-ASC schwannoma gene therapy to clinical trials. We determine the minimum effective dose of AAV1-P0-hASC required to induce an anti-tumor effect in the xenograft human-schwannoma model. We also show that the presence of preexisting AAV1 immunity does not alter the antitumor efficacy of AAV-P0-mASC in a syngeneic mouse schwannoma model. Furthermore, the maximum deliverable intratumoral dose of AAV1-P0-ASC was not associated with neuronal toxicity in immunocompetent mice. Taken together, these safety and efficacy data support the translation of the AAV1-P0-ASC schwannoma gene therapy strategy to clinical trials.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Neurilemoma/genética , Neurilemoma/terapia , Animais , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Imuno-Histoquímica , Camundongos , Transdução Genética , Transgenes , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Ann Clin Transl Neurol ; 8(7): 1508-1514, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34053190

RESUMO

Schwannomas are benign neoplasms that can cause gain- and loss-of-function neurological phenotypes, including severe, intractable pain. To investigate the molecular mechanisms underlying schwannoma-associated pain we compared the RNA sequencing profile of painful and non-painful schwannomas from NF2 patients. Distinct segregation of painful and non-painful tumors by gene expression patterns was observed. Differential expression analysis showed the upregulation of fibroblast growth factor 7 (FGF7) in painful schwannomas. Behavioral support for this finding was observed using a xenograft human NF2-schwannoma model in nude mice. In this model, over-expression of FGF7 in intra-sciatically implanted NF2 tumor cells generated pain behavior compared with controls.


Assuntos
Fator 7 de Crescimento de Fibroblastos/genética , Neurilemoma/genética , Neurofibromatose 2/genética , Dor/genética , Análise de Sequência de RNA/métodos , Transcriptoma/genética , Animais , Linhagem Celular Tumoral , Feminino , Fator 7 de Crescimento de Fibroblastos/biossíntese , Humanos , Masculino , Camundongos , Camundongos Nus , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurofibromatose 2/metabolismo , Neurofibromatose 2/patologia , Dor/metabolismo , Dor/patologia , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
Cancer Gene Ther ; 27(3-4): 265, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31056550

RESUMO

The original version of this Article contained an error in the spelling of the author Ahmed Abdelanabi, which was incorrectly given as Abdelanabi Ahmed. This has now been corrected in both the PDF and HTML versions of the Article.

7.
Hear Res ; 381: 107770, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31430634

RESUMO

Vestibular schwannoma (VS) is the fourth most common intracranial tumor, arising from neoplastic Schwann cells of the vestibular nerve and often causing debilitating sensorineural hearing loss (SNHL) and tinnitus. Previous research suggests that the abnormal upregulation of inflammatory pathways plays a highly significant, though infrequently described role in VS pathobiology, and that VS-associated SNHL is due not only to mechanical compression of the auditory nerve but also to differences in the intrinsic biology of these tumors. We hypothesize that patients who present with poor hearing associated with VS experience a more robust inflammatory response to this tumor than VS patients who present with good hearing. To investigate this hypothesis, we conducted a comprehensive pathway analysis using gene expression data from the largest meta-analysis of vestibular schwannoma microarray data, comprising 80 tumors and 16 healthy peripheral nerves. We identified the NLRP3 inflammasome as a novel target worthy of further exploration in VS research and validated this finding at the gene and protein expression level in human VS tissue using qRT-PCR and immunohistochemistry. To date, NLRP3 inflammasome activation has not been reported in VS, and this finding may represent a new and potentially significant therapeutic avenue. Notably, after analysis of 30 VSs, we observe that overexpression of key components of the NLRP3 inflammasome is preferentially associated with tumors that produce increased hearing loss in VS patients. Therefore, therapeutic development for VS should include considerations for minimizing NLRP3-associated inflammation to best preserve hearing.


Assuntos
Perda Auditiva Neurossensorial/etiologia , Audição , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neuroma Acústico/complicações , Nervo Vestibulococlear/metabolismo , Estudos de Casos e Controles , Regulação da Expressão Gênica , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neuroma Acústico/patologia , Nervo Vestibulococlear/patologia , Nervo Vestibulococlear/fisiopatologia
8.
J Neurosci Methods ; 323: 77-81, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31125589

RESUMO

BACKGROUND: Schwannomas are peripheral nerve sheath tumors composed entirely of Schwann-lineage cells that cause pain and sensory-motor dysfunction through compression of peripheral nerves, the spinal cord, and/or the brain stem. Treatment of schwannoma is largely limited to resection which itself has limited value. The goal of this study is to establish a technique to identify the most efficient and tissue-specific promoter for use in a schwannoma gene therapy construct. NEW METHOD: This work involves transfection of schwannoma cells with adeno-associated viral vector plasmids expressing GFP under different myelin cell specific promoters. The transfected cells were evaluated for green fluorescence intensity in vitro, and in vivo after implantation into sciatic nerves of nude mice. RESULTS: Our data demonstrate that myelin protein zero (MPZ, P0) and peripheral myelin protein 22 (PMP22) promoters produce greater GFP expression in schwannoma cell lines than myelin basic protein (MBP) promoter. In vitro, P0 promoter activity in schwannoma cell lines was shown to be less active than the cytomegalovirus and chicken ß-actin (CBA) promoter. However, we did not observe any significant difference between the activity of the CBA and P0 promoters in a xenograft schwannoma model. COMPARISON WITH EXISTING METHODS(S): We show here the influence of the peripheral nerve microenvironment on promoter efficacy in expressing transgenes using simple transfection by lipofection followed by prompt implantation of the transfected cells into the sciatic nerve of nude mice. CONCLUSIONS: We demonstrate that of the myelin specific promoters evaluated, P0 is optimal for driving expression of transgenes in schwannoma cells.


Assuntos
Neoplasias do Sistema Nervoso Central/terapia , Terapia Genética , Proteína Básica da Mielina , Proteína P0 da Mielina/metabolismo , Proteínas da Mielina , Neurilemoma/terapia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Proteína Básica da Mielina/metabolismo
9.
Neuro Oncol ; 21(7): 854-866, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-30977509

RESUMO

BACKGROUND: We evaluated apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) as a schwannoma tumor suppressor and explored its utilization in a schwannoma gene therapy strategy that may be translated to clinical use. METHODS: ASC protein expression and mRNA level were assessed in human schwannoma by immunohistochemistry and quantitative PCR, respectively. Methylation- specific PCR was used to assess ASC promoter methylation. The effect of ASC overexpression in schwannoma cells was evaluated through ATP-based viability, lactate dehydrogenase release, and apoptosis staining. Western blotting and colorimetric assay were used to test the effect of ASC overexpression on endogenous pro-apoptotic pathways. Bioluminescence imaging, behavioral testing, and immunohistochemistry in human xenograft and murine allograft schwannoma models were used to examine the efficacy and toxicity of intratumoral injection of adeno-associated virus (AAV) vector encoding ASC. RESULTS: ASC expression was suppressed via promoter methylation in over 80% of the human schwannomas tested. ASC overexpression in schwannoma cells results in cell death and is associated with activation of endogenous caspase-9, caspase-3, and upregulation of BH3 interacting-domain death agonist. In a human xenograft schwannoma model, AAV1-mediated ASC delivery reduced tumor growth and resolved tumor-associated pain without detectable toxicity, and tumor control was associated with reduced Ki67 mitotic index and increased tumor-cell apoptosis. Efficacy of this schwannoma gene therapy strategy was confirmed in a murine schwannoma model. CONCLUSION: We have identified ASC as a putative schwannoma tumor suppressor with high potential clinical utility for schwannoma gene therapy and generated a vector that treats schwannomas via a novel mechanism that does not overlap with current treatments.


Assuntos
Apoptose , Biomarcadores Tumorais/genética , Proteínas Adaptadoras de Sinalização CARD/administração & dosagem , Dor do Câncer/prevenção & controle , Terapia Genética , Neurilemoma/terapia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Dor do Câncer/etiologia , Proliferação de Células , Metilação de DNA , Dependovirus/genética , Humanos , Masculino , Camundongos , Neurilemoma/genética , Neurilemoma/patologia , Prognóstico , Regiões Promotoras Genéticas , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Gene Ther ; 26(9-10): 259-267, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30622323

RESUMO

Schwannomas are peripheral nerve sheath tumors associated with three genetically distinct disease entities, namely sporadic schwannoma, neurofibromatosis type-2, and schwannomatosis. Schwannomas are associated with severe disability and in cases lead to death. The primary treatment is operative resection that itself can cause neurologic damage and is at times contra-indicated due to tumor location. Given their homogenous Schwann-lineage cellular composition, schwannomas are appealing targets for gene therapy. In the present study, we have generated an adeno-associated serotype 1 virus (AAV1)-based vector delivering N-terminal of the pyroptotic gene Gasdermin-D; (GSDMDNterm) under the control of the Schwann-cell specific promoter, P0. we have demonstrated that AAV1-P0-GSDMDNterm injection into intra-sciatic schwannomas reduces the growth of these tumors and resolves tumor-associated pain without causing neurologic damage. This AAV1-P0-GSDMDNterm vector holds promise for clinical treatment of schwannomas via direct intra-tumoral injection.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neurilemoma/genética , Proteínas de Ligação a Fosfato/genética , Animais , Biomarcadores , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intralesionais , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Atividade Motora/genética , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurilemoma/terapia , Neurônios/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Transdução de Sinais , Transdução Genética , Transgenes , Resultado do Tratamento , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Control Release ; 273: 99-107, 2018 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-29289570

RESUMO

Integration-deficient lentiviral vectors (IDLVs) are promising gene delivery tools that retain the high transduction efficiency of standard lentiviral vectors, yet fail to integrate as proviruses and are instead converted into episomal circles. These episomes are metabolically stable and support long-term expression of transgenes in non-dividing cells, exhibiting a decreased risk of insertional mutagenesis. We have embarked on an extensive study to compare the transduction efficiency of IDLVs pseudotyped with different envelopes (vesicular stomatitis, Rabies, Mokola and Ross River viral envelopes) and self-complementary adeno-associated viral vectors, serotype-9 (scAAV-9) in spinal cord tissues after intraspinal injection of mouse embryos (E16). Our results indicate that IDLVs can transduce motor neurons (MNs) at extremely high efficiency regardless of the envelope pseudotype while scAAV9 mediates gene delivery to ~40% of spinal cord motor neurons, with other non-neuronal cells also transduced. Long-term expression studies revealed stable gene expression at 7months post-injection. Taken together, the results of this study indicate that IDLVs may be efficient tools for in utero cord transduction in therapeutic strategies such as for treatment of inherited early childhood neurodegenerative diseases.


Assuntos
Técnicas de Transferência de Genes , Lentivirus , Neurônios Motores , Medula Espinal , Adenoviridae , Animais , Feminino , Feto , Células HEK293 , Humanos , Injeções , Camundongos , Gravidez , Útero , Proteínas do Envelope Viral
12.
Hum Gene Ther ; 25(5): 428-42, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24483952

RESUMO

Nonintegrating gene delivery vectors have an improved safety profile compared with integrating vectors, but transgene retention is problematic as nonreplicating episomes are progressively and rapidly diluted out through cell division. We have developed an integration-deficient lentiviral vector (IDLV) system generating mitotically stable episomes capable of long-term transgene expression. We found that a transient cell cycle arrest at the time of transduction with IDLVs resulted in 13-45% of Chinese hamster ovary (CHO) cells expressing the transgene for over 100 cell generations in the absence of selection. The use of a scaffold/matrix attachment region did not result in improved episomal retention in this system, and episomes did not form after transduction with adeno-associated viral or minicircle vectors under the same conditions. Investigations into the episomal status of the vector genome using (1) linear amplification-mediated polymerase chain reaction followed by deep sequencing of vector-genome junctions, (2) Southern blotting, and (3) fluorescent in situ hybridization strongly suggest that the vector is not integrated in the vast majority of cells. In conclusion, we have developed an IDLV procedure generating mitotically stable episomes capable of long-term transgene expression. The application of this approach to stem cell populations could significantly improve the safety profile of a range of stem and progenitor cell gene therapies.


Assuntos
Expressão Gênica , Vetores Genéticos/metabolismo , Lentivirus/genética , Mitose , Plasmídeos/genética , Transgenes/genética , Integração Viral , Animais , Células CHO , Pontos de Checagem do Ciclo Celular , Núcleo Celular/metabolismo , Células Clonais , Cricetinae , Cricetulus , DNA Circular/metabolismo , Dependovirus/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Regiões de Interação com a Matriz , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Origem de Replicação/genética , Fatores de Tempo , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA