Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Leukemia ; 31(12): 2791-2798, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28439108

RESUMO

MV-NIS is an Edmonston lineage oncolytic measles virus expressing the human sodium iodide symporter-a means for monitoring by non-invasive imaging of radioiodine. Patients with relapsed, refractory myeloma who had explored all other treatment options were eligible for this Phase I trial. Cohort 1 was treated with intravenous MV-NIS, and Cohort 2 received cyclophosphamide 2 days prior to MV-NIS. Thirty-two patients were treated. Cohort 1 initially enrolled to four dose levels without reaching maximum tolerated dose (MTD) and subsequently to two higher dose levels when improved virus manufacture technology made it possible. MTD was not reached in Cohort 1, and TCID50 1011 is the dose being used in a Phase II trial of single agent MV-NIS. Grade 3-4 adverse events in both cohorts at all dose levels were: neutropenia (n=9); leukocyte count decreased (n=5); thrombocytopenia (n=2); and CD4 lymphocytes decreased, anemia and lymphopenia (each n=1). MV-N RNA sequences were amplified from gargle specimens, blood and urine. 123I scans were positive in eight patients. One patient achieved a complete response; transient drops in serum free light chains were seen in other patients. MV-NIS is capable of replicating before being cleared by the immune system. Oncolytic viruses offer a promising new modality for the targeted infection and destruction of disseminated myeloma.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Vírus do Sarampo/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Simportadores/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Feminino , Engenharia Genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mieloma Múltiplo/diagnóstico , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Recidiva , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Distribuição Tecidual , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Eur J Surg Oncol ; 42(7): 1024-34, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27179924

RESUMO

BACKGROUND: This work was to evaluate the efficacy and safety of cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) with lobaplatin and docetaxel to treat peritoneal carcinomatosis (PC) from gastric cancer (GC). METHODS: A total of 50 consecutive GC PC patients treated by 52 CRS+HIPEC procedures with lobaplatin 50 mg/m(2) and docetaxel 60 mg/m(2) in 6000 mL of normal saline at (43 ± 0.5) °C for 60 min. The primary endpoint was overall survival (OS), and the secondary endpoints were perioperative safety profiles. RESULTS: At the median follow-up of 22.5 (range, 5.1-50.7) months, the median OS was 14.3 (95% CI 7.6-21.0) months, and the 1-, 2-, and 3-year survival rates were 58%, 40%, and 32%, respectively. Mortality and serious adverse event (grade 3-5) morbidity rates in postoperative 30 days were 0.0% and 23.1%, respectively. Univariate analysis identified 4 parameters with significant effects on OS: completeness of cytoreduction (CC) 0-1, normal (N) the preoperative tumor markers level (TM), adjuvant chemotherapy ≥6 cycles, and peritoneal cancer index ≤20. However, multivariate analysis identified CC0-1, perioperative TM (N), adjuvant chemotherapy ≥6 cycles as the independent predictor for better survival. CONCLUSIONS: CRS+HIPEC with lobaplatin and docetaxel to treat selected GC PC could improve OS, with acceptable perioperative safety.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/cirurgia , Quimioterapia do Câncer por Perfusão Regional , Procedimentos Cirúrgicos de Citorredução , Hipertermia Induzida , Neoplasias Primárias Múltiplas/tratamento farmacológico , Neoplasias Primárias Múltiplas/cirurgia , Neoplasias Peritoneais/tratamento farmacológico , Neoplasias Peritoneais/cirurgia , Neoplasias Gástricas/patologia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma/mortalidade , Carcinoma/secundário , China , Ciclobutanos/administração & dosagem , Bases de Dados Factuais , Docetaxel , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Neoplasias Primárias Múltiplas/mortalidade , Compostos Organoplatínicos/administração & dosagem , Neoplasias Peritoneais/mortalidade , Neoplasias Peritoneais/secundário , Prognóstico , Estudos Retrospectivos , Tamanho da Amostra , Taxoides/administração & dosagem , Resultado do Tratamento
3.
Cancer Gene Ther ; 21(6): 256-60, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24874841

RESUMO

The relationship between ligand-receptor affinity and antitumor potency of an oncolytic virus was investigated using a panel of six HER2/neu (HER2)-targeted measles viruses (MVs) displaying single-chain antibodies (scFv) that bind to the same epitope on HER2, but with affinities ranging from 10(-6) to 10(-11) M. All viruses were able to infect SKOV3ip.1 human ovarian cancer cells in vitro, but only the high-affinity MV (Kd≥10(-8) M) induced cytopathic effects of syncytia formation in the cell monolayers. In contrast, all six viruses were therapeutically active in vivo against orthotopic human ovarian SKOV3ip.1 tumor xenografts in athymic mice compared with saline-treated controls. The oncolytic activities of MV displaying the high-affinity scFv (Kd=10(-9), 10(-10), 10(-11) M) were not significantly superior to MV displaying scFv with Kd of 10(-8) M or less. Results from this study suggest that increasing the receptor affinity of the attachment protein of an oncolytic MV has minimal impact on its in vivo efficacy against a tumor that expresses the targeted receptor.


Assuntos
Vírus do Sarampo/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Anticorpos de Cadeia Única/metabolismo , Animais , Modelos Animais de Doenças , Epitopos/metabolismo , Feminino , Humanos , Injeções Intraperitoneais , Camundongos Nus , Vírus Oncolíticos/patogenicidade , Neoplasias Ovarianas/virologia , Receptor ErbB-2/metabolismo , Esferoides Celulares/virologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Radiol ; 69(2): 123-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24315547

RESUMO

AIM: To determine the features of caesarean scar ectopic pregnancy (CSP) by using magnetic resonance imaging (MRI) in the first trimester. MATERIALS AND METHODS: Thirty-nine women with CSP in the first trimester were initially selected and underwent transvaginal ultrasound and MRI examinations. Thirty-five patients with CSP were finally included in this study. RESULTS: The CSPs were categorized into three groups: type I, in which a thin-walled diverticulum is present at the caesarean section scar (CSS) defect and the gestational sac (GS) is embedded in the diverticulum; type II, in which a thin-walled diverticulum is present at the CSS defect and the GS is partially embedded in the diverticulum; type III, in which a niche is present in the CSS defect and the GS is mainly embedded in the isthmus. Types I, II, and III CSP occurred in 40, 46, and 14% of the women, respectively. There was no significant difference between the three types in the minimum thickness of the CSS defect. In types I and II, there was a positive correlation in the maximum inlet diameter of the CSS defect and the approximate area of the GS. CONCLUSION: Using MRI, most CSPs present as a diverticulum at the CSS defect, and the CSS defect becomes weaker with the growth of the GS. MRI showed detailed features of the CSP.


Assuntos
Cesárea/efeitos adversos , Cicatriz/patologia , Imageamento por Ressonância Magnética/métodos , Primeiro Trimestre da Gravidez , Gravidez Ectópica/diagnóstico , Adulto , Cicatriz/complicações , Cicatriz/etiologia , Diagnóstico Diferencial , Feminino , Humanos , Processamento de Imagem Assistida por Computador/métodos , Gravidez , Adulto Jovem
5.
Cancer Gene Ther ; 20(11): 616-21, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24176894

RESUMO

Vesicular stomatitis virus (VSV) is neuropathogenic in rodents but can be attenuated 50-fold by engineering the mouse interferon-beta (IFN-ß) gene into its genome. Intravenously administered VSVs encoding IFN-ß have potent activity against subcutaneous tumors in the 5TGM1 mouse myeloma model, without attendant neurotoxicity. However, when 5TGM1 tumor cells were seeded intravenously, virus-treated mice with advanced myeloma developed clinical signs suggestive of meningoencephalitis. Co-administration of a known active antimyeloma agent did not prolong survival, further suggesting that deaths were due to viral toxicity, not tumor burden. Histological analysis revealed that systemically administered 5TGM1 cells seed to the CNS, forming meningeal tumor deposits, and that VSV infects and destroys these tumors. Death is presumably a consequence of meningeal damage and/or direct transmission of virus to adjacent neural tissue. In light of these studies, extreme caution is warranted in clinical testing of attenuated VSVs, particularly in patients with CNS tumor deposits.


Assuntos
Neoplasias Meníngeas/terapia , Neoplasias Meníngeas/virologia , Mieloma Múltiplo/terapia , Mieloma Múltiplo/virologia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/genética , Vesiculovirus/genética , Animais , Modelos Animais de Doenças , Feminino , Interferon beta/genética , Neoplasias Meníngeas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/patologia
6.
Cancer Gene Ther ; 20(11): 638-41, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24030210

RESUMO

Sodium iodide symporter (NIS) reporter gene imaging is an excellent technology for noninvasive cell fate determination in living animals unless the NIS-transduced cells reside in perigastric organs such as the spleen, liver, diaphragm, omentum, pancreas, perigastric lymph nodes or perigastric tumor deposits. Here we report that orally administered barium sulfate enhances CT definition of the stomach, masks background gamma ray emissions from the stomach and enhances signal detection from radiotracer uptake in NIS-transduced organs.


Assuntos
Sulfato de Bário , Meios de Contraste , Genes Reporter/efeitos dos fármacos , Radioisótopos do Iodo , Simportadores/genética , Animais , Sulfato de Bário/administração & dosagem , Linhagem Celular Tumoral , Meios de Contraste/administração & dosagem , Feminino , Técnicas de Transferência de Genes , Xenoenxertos , Humanos , Radioisótopos do Iodo/administração & dosagem , Camundongos , Camundongos Nus , Imagem Multimodal/métodos , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/patologia , Estômago/anatomia & histologia , Estômago/diagnóstico por imagem , Simportadores/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Tomografia Computadorizada por Raios X/métodos
7.
Cancer Gene Ther ; 20(10): 582-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24030211

RESUMO

Oncolytic vesicular stomatitis virus (VSV) has potent antitumor activity but some cancer cells are resistant to VSV killing, either constitutively or due to type I interferon (IFN) inducing an antiviral state in the cells. Here, we evaluated VSV oncolysis of a panel of human head and neck cancer cells and showed that VSV resistance in SCC25 and SCC15 cells could be reversed with Janus kinase (JAK) 1/2 inhibitors (JAK inhibitor I and ruxolitinib). Pre-treatment of cells with JAK1/2 inhibitors before or in conjunction with VSV enhanced viral infection, spread and progeny yield (100- to 1000-fold increase). In contrast, inhibitors of histone deacetylase (LBH589), phosphatidylinositol 3-kinase (GDC-0941, LY294002), mammalian target of rapamycin (rapamycin) or signal transducer and activator of transcription 3 (STAT3 inhibitor VII) were ineffective. Compared with VSV-sensitive SW579 cells, IFNα/ß responsive antiviral genes (IRF-9, IRF-7, OAS1 but not MxA) are constitutively expressed in SCC25 cells. Pretreatment with JAK inhibitors reduced mRNA levels of these genes, increasing VSV expression in the cells. Interestingly, 1 h of drug exposure was sufficient to reverse SCC25 resistance to VSV and was still effective if virus was added 24 h later. Overall, we show here that JAK inhibitor I and ruxolitinib (Jakafi) can reverse resistance to VSV, supporting the rationale to incorporate JAK1/2 inhibitors in future VSV virotherapy trials.


Assuntos
Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/virologia , Neoplasias de Cabeça e Pescoço/terapia , Neoplasias de Cabeça e Pescoço/virologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Terapia Viral Oncolítica/métodos , Inibidores de Proteínas Quinases/farmacologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/imunologia , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/imunologia , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Janus Quinase 1/biossíntese , Janus Quinase 1/genética , Janus Quinase 2/biossíntese , Janus Quinase 2/genética , Nitrilas , Pirazóis/farmacologia , Pirimidinas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo , Replicação Viral
8.
Cancer Gene Ther ; 20(3): 141-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23370333

RESUMO

We provide an overview of the latest developments in cancer gene therapy--from the bench to early-stage clinical trials. We describe the most recent work of worldwide teams including experienced scientists and clinicians, reflecting the recent emergence of gene therapy from the 'Valley of Death'. The treatment efficacy of clinical gene therapy has now been shown in a number of diseases including cancer and we are observing a renewed interest by big pharmaceutical and biotechnology companies most obviously demonstrated by Amgen's acquisition of Biovex for up to USD$1 billion. There is an opportunity to be cautiously hopeful regarding the future of gene therapy in the clinic and we review here some of the most recent progress in the field.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Animais , Biomarcadores Tumorais/genética , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Imunoterapia , Terapia de Alvo Molecular , Medicina de Precisão , Interferência de RNA
9.
Gene Ther ; 20(3): 255-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22476202

RESUMO

Oncolytic viruses can be neutralized in the bloodstream by antiviral antibodies whose titers increase progressively with each exposure, resulting in faster virus inactivation and further reductions in efficacy with each successive dose. A single dose of cyclophosphamide (CPA) at 370 mg m(-2) was not sufficient to control the primary antiviral immune responses in mice, squirrel monkeys and humans. We therefore tested clinically approved multidose CPA regimens, which are known to kill proliferating lymphocytes, to determine if more intensive CPA therapy can more effectively suppress antiviral antibody responses during virotherapy. In virus-susceptible mice, primary antibody responses to intravenously (i.v.) administered oncolytic measles virus (MV) or vesicular stomatitis virus (VSV) were partially or completely suppressed, respectively, by oral (1 mg × 8 days) or systemic (3 mg × 4 days) CPA regimens initiated 1 day before virus. When MV- or VSV-immune mice were re-challenged with the respective viruses and concurrently treated with four daily systemic doses of CPA, their anamnestic antibody responses were completely suppressed and antiviral antibody titers fell significantly below pre-booster levels. We conclude that the CPA regimen of four daily doses at 370 mg m(-2) should be evaluated clinically with i.v. virotherapy to control the antiviral antibody response and facilitate effective repeat dosing.


Assuntos
Ciclofosfamida/farmacologia , Imunidade Humoral/efeitos dos fármacos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Anticorpos Antivirais/imunologia , Cricetinae , Ciclofosfamida/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Imunidade Humoral/imunologia , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Sarampo/imunologia , Sarampo/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Fatores de Tempo , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/fisiologia
10.
Cancer Gene Ther ; 19(9): 659-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22790962

RESUMO

Anaplastic thyroid cancer is an extremely aggressive disease resistant to radioiodine treatment because of loss of sodium iodide symporter (NIS) expression. To enhance prognosis of this fatal cancer, we validated the preclinical efficacy of measles virus (MV)-NIS, the vaccine strain of the oncolytic MV (MV-Edm), modified to include the NIS gene. Western blotting analysis confirmed that a panel of eight anaplastic thyroid cancer (ATC)-derived cell lines do not express NIS protein, but do express CD46, the MV receptor. In vitro cell death assays and in vivo xenograft studies demonstrate the oncolytic efficacy of MV-NIS in BHT-101 and KTC-3, ATC-derived cell lines. Radioactive iodine uptake along with single-photon emission computed tomography (SPECT)-computed tomography imaging of KTC-3 xenografts after (99)Tc(m) administration confirmed NIS expression in vitro and in vivo, respectively, after virus treatment. Adjuvant administration of RAI, to MV-NIS-treated KTC-3 tumors showed a trend for increased tumor cell killing. As current treatment for ATC is only palliative, and MV-NIS is currently Food and Drug Administration approved for human clinical trials in myeloma, our data indicate that targeting ATC with MV-NIS could prove to be a novel therapeutic strategy for effective treatment of iodine-resistant ATC and will expedite its testing in clinical trials for this aggressive disease.


Assuntos
Iodo/metabolismo , Vírus do Sarampo/metabolismo , Vírus Oncolíticos/metabolismo , Simportadores/uso terapêutico , Neoplasias da Glândula Tireoide/terapia , Animais , Western Blotting , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Terapia Genética/métodos , Humanos , Radioisótopos do Iodo/metabolismo , Radioisótopos do Iodo/uso terapêutico , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Nus , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Receptores Virais/metabolismo , Simportadores/genética , Simportadores/metabolismo , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Leukemia ; 26(8): 1870-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22425894

RESUMO

Current therapy for multiple myeloma is complex and prolonged. Antimyeloma drugs are combined in induction, consolidation and/or maintenance protocols to destroy bulky disease, then suppress or eradicate residual disease. Oncolytic viruses have the potential to mediate both tumor debulking and residual disease elimination, but this curative paradigm remains unproven. Here, we engineered an oncolytic vesicular stomatitis virus to minimize its neurotoxicity, enhance induction of antimyeloma immunity and facilitate noninvasive monitoring of its intratumoral spread. Using high-resolution imaging, autoradiography and immunohistochemistry, we demonstrate that the intravenously administered virus extravasates from tumor blood vessels in immunocompetent myeloma-bearing mice, nucleating multiple intratumoral infectious centers that expand rapidly and necrose at their centers, ultimately coalescing to cause extensive tumor destruction. This oncolytic tumor debulking phase lasts only for 72 h after virus administration, and is completed before antiviral antibodies become detectable in the bloodstream. Antimyeloma T cells, cross-primed as the virus-infected cells provoke an antiviral immune response, then eliminate residual uninfected myeloma cells. The study establishes a curative oncolytic paradigm for multiple myeloma where direct tumor debulking and immune eradication of minimal disease are mediated by a single intravenous dose of a single therapeutic agent. Clinical translation is underway.


Assuntos
Vetores Genéticos/administração & dosagem , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Animais , Linhagem Celular Tumoral , Cricetinae , Terapia Genética , Vetores Genéticos/genética , Interferon beta/genética , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Vírus Oncolíticos/genética , Simportadores/genética , Simportadores/metabolismo , Transplante Isogênico , Vírus da Estomatite Vesicular Indiana/genética
12.
Cancer Gene Ther ; 19(3): 202-11, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22116376

RESUMO

Off-target binding or vector sequestration can significantly limit the efficiency of systemic virotherapy. We report here that systemically administered oncolytic measles virus (MV) was rapidly sequestered by the mononuclear phagocytic system (MPS) of the liver and spleen in measles receptor CD46-positive and CD46-negative mice. Since scavenger receptors on Kupffer cells are responsible for the elimination of blood-borne pathogens, we investigated here if MV uptake was mediated by scavenger receptors on Kupffer cells. Pretreatment of cells with poly(I), a scavenger receptor ligand, reduced MV expression by 99% in murine (J774A.1) macrophages and by 50% in human (THP-1) macrophages. Pre-dosing of mice with poly(I) reduced MPS sequestration of MV and increased circulating levels of MV by 4 to 15-folds at 2 min post virus administration. Circulating virus was still detectable 30 min post infusion in mice pre-dosed with poly(I) whereas no detectable MV was found at 5-10 min post infusion if mice did not receive poly(I). MPS blockade by poly(I) enhanced virus delivery to human ovarian SKOV3ip.1 and myeloma KAS6/1 xenografts in mice. Higher gene expression and improved control of tumor growth was noted early post therapy. Based on these results, incorporation of MPS blockade into MV treatment regimens is warranted.


Assuntos
Vírus do Sarampo/fisiologia , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Ovarianas/terapia , Poli I/farmacologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Humanos , Imuno-Histoquímica , Fígado/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Camundongos , Camundongos Nus , Camundongos Transgênicos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/virologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/virologia , Baço/virologia , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Gene Ther ; 17(8): 550-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20379224

RESUMO

Mesothelioma usually leads to death within 8-14 months of diagnosis. To increase the potency of oncolytic measles viruses (MVs) for mesothelioma therapy, we inserted the interferon beta (IFNbeta) gene alone or with the human thyroidal sodium iodide symporter (NIS) gene into attenuated MV of the Edmonston lineage. The corresponding mouse IFNbeta (mIFNbeta) viruses, MV-mIFNbeta and MV-mIFNbeta-NIS, successfully propagated in human mesothelioma cells, leading to intercellular fusion and cell death. High levels of mIFNbeta were detected in the supernatants of the infected cells, and radioiodine uptake was substantial in the cells infected with MV-mIFNbeta-NIS. MV with mIFNbeta expression triggered CD68-positive immune cell infiltration 2-4 times higher than MV-GFP injected into the tumor site. The numbers of CD31-positive vascular endothelial cells within the tumor were decreased at day 7 after intratumoral injection of MV-mIFNbeta or MV-mIFNbeta-NIS, but not after MV-GFP and PBS administration. Immunohistochemical analysis showed that MV-mIFNbeta changed the microenvironment of the mesothelioma by increasing innate immune cell infiltration and inhibiting tumor angiogenesis. Oncolytic MVs coding for IFNbeta effectively retarded growth of human mesotheliomas and prolonged survival time in several mesothelioma tumor models. The results suggest that oncolytic MVs that code for IFNbeta and NIS will be potent and versatile agents for the treatment of human mesothelioma.


Assuntos
Interferon beta/genética , Vírus do Sarampo/fisiologia , Mesotelioma/terapia , Terapia Viral Oncolítica/métodos , Simportadores/genética , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Humanos , Interferon beta/biossíntese , Radioisótopos do Iodo , Vírus do Sarampo/genética , Mesotelioma/diagnóstico por imagem , Mesotelioma/virologia , Camundongos , Camundongos Nus , Camundongos SCID , Plasmídeos/genética , Simportadores/biossíntese , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Gene Ther ; 16(12): 873-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19498461

RESUMO

Replication-competent viruses are being tested as tumor therapy agents. The fundamental premise of this therapy is the selective infection of the tumor cell population with the amplification of the virus. Spread of the virus in the tumor ultimately should lead to eradication of the cancer. Tumor virotherapy is unlike any other form of cancer therapy as the outcome depends on the dynamics that emerge from the interaction between the virus and tumor cell populations both of which change in time. We explore these interactions using a model that captures the salient biological features of this system in combination with in vivo data. Our results show that various therapeutic outcomes are possible ranging from tumor eradication to oscillatory behavior. Data from in vivo studies support these conclusions and validate our modeling approach. Such realistic models can be used to understand experimental observations, explore alternative therapeutic scenarios and develop techniques to optimize therapy.


Assuntos
Vacina contra Sarampo/uso terapêutico , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica/métodos , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Camundongos , Camundongos SCID , Modelos Biológicos , Modelos Estatísticos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/virologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Curr Top Microbiol Immunol ; 330: 213-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19203112

RESUMO

Measles virus offers an ideal platform from which to build a new generation of safe, effective oncolytic viruses. Occasional so-called spontaneous tumor regressions have occurred during natural measles infections, but common tumors do not express SLAM, the wild-type MV receptor, and are therefore not susceptible to the virus. Serendipitously, attenuated vaccine strains of measles virus have adapted to use CD46, a regulator of complement activation that is expressed in higher abundance on human tumor cells than on their nontransformed counterparts. For this reason, attenuated measles viruses are potent and selective oncolytic agents showing impressive antitumor activity in mouse xenograft models. The viruses can be engineered to enhance their tumor specificity, increase their antitumor potency, and facilitate noninvasive in vivo monitoring of their spread. A major impediment to the successful deployment of oncolytic measles viruses as anticancer agents is the high prevalence of preexisting anti-measles immunity, which impedes bloodstream delivery and curtails intratumoral virus spread. It is hoped that these problems can be addressed by delivering the virus inside measles-infected cell carriers and/or by concomitant administration of immunosuppressive drugs. From a safety perspective, population immunity provides an excellent defense against measles spread from patient to carers and, in 50 years of human experience, reversion of attenuated measles to a wild-type pathogenic phenotype has not been observed. Clinical trials testing oncolytic measles viruses as an experimental cancer therapy are currently underway.


Assuntos
Vírus do Sarampo/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Animais , Engenharia Genética , Terapia Genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/imunologia , Camundongos , Neoplasias/imunologia , Vírus Oncolíticos/genética
16.
Clin Pharmacol Ther ; 82(6): 700-10, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17971816

RESUMO

MV-NIS is an oncolytic measles virus encoding the human thyroidal sodium iodide symporter (NIS). Here, we report the results of preclinical pharmacology and toxicology studies conducted in support of our clinical protocol "Phase I Trial of Systemic Administration of Edmonston Strain of Measles Virus, Genetically Engineered to Express NIS, with or without Cyclophosphamide, in Patients with Recurrent or Refractory Multiple Myeloma." Dose-response studies in the KAS-6/1 myeloma xenograft model demonstrated a minimum effective dose of 4 x 10(6) TCID50 (tissue culture infectious dose 50)/kg. Toxicity studies in measles-naive squirrel monkeys and measles-susceptible transgenic mice were negative at intravenous doses up to 10(8) and 4 x 10(8) TCID50/kg, respectively. Abundant viral mRNA, maximal on day 8, was detected in cheek swabs of squirrel monkeys, more so after pretreatment with cyclophosphamide. On the basis of these data, the safe starting dose of MV-NIS for our clinical protocol was set at 1-2 x 10(4) TCID50/kg (10(6) TCID50 per patient).


Assuntos
Antineoplásicos/farmacologia , Ciclofosfamida/farmacologia , Vírus do Sarampo , Mieloma Múltiplo/tratamento farmacológico , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Simportadores/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Ciclofosfamida/administração & dosagem , Feminino , Humanos , Injeções Intravenosas , Vírus do Sarampo/genética , Vírus do Sarampo/isolamento & purificação , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Camundongos Transgênicos , Terapia Viral Oncolítica/efeitos adversos , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saimiri , Simportadores/administração & dosagem , Transplante Heterólogo
17.
Gene Ther ; 14(4): 324-33, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17051248

RESUMO

Neutralizing antiviral antibodies (Abs) can hinder systemic virotherapy. Here, we used activated T cells as carriers to deliver oncolytic measles viruses (MV) to multiple myeloma xenografts in the presence of anti-MV antibodies (Abs). Virus-infected T cells expressing measles H/F fusogenic envelope glycoproteins could efficiently transfer MV infection by heterofusion, even after exposure to virus-inactivating anti-MV antisera. Severe-combined immunodeficiency (SCID) mice bearing subcutaneous or disseminated human myeloma xenografts were given MV-luciferase (MV-Luc) or MV-Luc-infected T cells intravenously. Indium111 labeling indicated that 1-2% of the virus-infected T cells trafficked to tumors. Preinfected T cells fused with tumor cells in vivo and transferred MV-Luc to tumor xenografts where intratumoral viral spread was monitored non-invasively using bioluminescent imaging. In mice passively immunized with high titers of measles immune serum, intravenous virus and cell delivery were both inhibited. Decreasing the amount of measles immune serum given to mice permitted tumor infection by virus-infected T cells and cell-free virus. In conclusion, virus-loaded T cells may facilitate systemic measles virotherapy in the presence of antiviral Abs and they warrant further investigation as potential MV cell carriers.


Assuntos
Anticorpos Antivirais/imunologia , Vírus do Sarampo/imunologia , Sarampo/imunologia , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica/métodos , Linfócitos T/virologia , Animais , Feminino , Câmaras gama , Proteínas de Fluorescência Verde/genética , Humanos , Radioisótopos de Índio , Injeções , Luciferases/genética , Ativação Linfocitária , Vacina contra Sarampo/imunologia , Camundongos , Camundongos SCID , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/virologia , Transplante de Neoplasias , Testes de Neutralização , Compostos Radiofarmacêuticos , Linfócitos T/imunologia , Transdução Genética/métodos , Transplante Heterólogo
18.
Cancer Gene Ther ; 13(8): 732-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16543921

RESUMO

Because of their ability to replicate, the dose-response relationships of oncolytic viruses cannot easily be predicted. To better understand the pharmacokinetics of virotherapy in relation to viral dose and schedule, we administered MV-CEA intraperitoneally in an orthotopic mouse model of ovarian cancer. MV-CEA is an attenuated oncolytic measles virus engineered to express soluble human carcinoembryonic antigen (CEA), and the virus is currently undergoing phase I clinical testing in patients with ovarian cancer. Plasma CEA levels correlate with numbers of virus-infected tumor cells at a given time, and were used as a surrogate to monitor the profiles of viral gene expression over time. The antineoplastic activity of single- or multiple-dose MV-CEA was apparent over a wide range of virus doses (10(3)-10(8) TCID(50)), with little reduction in observed antitumor efficacy, even at the lowest tested dose. However, analysis of CEA profiles of treated mice was highly informative, illustrating the variability in virus kinetics at different dose levels. The highest doses of virus were associated with higher initial levels of tumor cell killing, but the final outcome of MV-CEA therapy at all dose levels was a partial equilibrium between virus and tumor, resulting in significant slowing of tumor growth and enhanced survival of the mice.


Assuntos
Vírus do Sarampo , Terapia Viral Oncolítica , Neoplasias Ovarianas/terapia , Animais , Antígeno Carcinoembrionário/biossíntese , Antígeno Carcinoembrionário/sangue , Relação Dose-Resposta Imunológica , Feminino , Humanos , Camundongos , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Carga Tumoral , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Gene Ther ; 12(1): 87-94, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15385953

RESUMO

In clinical trials with cancer patients, the safety of conditionally replicating adenoviruses (CRAds) has been good. However, marginal data are available on the persistence or antitumor efficacy of these agents. The oncolytic potency of CRAds is determined by their capacity for entering target cells. Consequently, we constructed a retargeted CRAd featuring a secreted marker protein, soluble human carcinoembryogenic antigen (hCEA), which can be measured in growth medium or plasma. We found that virus replication closely correlated with hCEA secretion both in vitro and in vivo. Further, antitumor efficacy and the persistence of the virus could be deduced from plasma hCEA levels. Finally, using in vivo bioluminescence imaging, we were able to detect effective tumor cell killing by the virus, which led to enhanced therapeutic efficacy.


Assuntos
Adenocarcinoma/terapia , Adenoviridae/genética , Antígeno Carcinoembrionário/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Neoplasias Ovarianas/terapia , Adenocarcinoma/sangue , Adenocarcinoma/virologia , Adenoviridae/fisiologia , Animais , Biomarcadores Tumorais/sangue , Antígeno Carcinoembrionário/sangue , Feminino , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos SCID , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/virologia , Resultado do Tratamento , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...