Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Aesthet Surg J ; 43(11): 1325-1333, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37265022

RESUMO

BACKGROUND: The incidence of seroma after abdominoplasty is accepted as approximately 10% (with a range) in the literature. Progressive tension sutures (PTS) have arisen as a means of reducing seroma, however there are conflicting data regarding their efficacy. OBJECTIVES: The primary aim of this study was to describe the incidence of postabdominoplasty seroma in the setting of drainless abdominoplasty with PTS. METHODS: A retrospective chart review was performed of all abdominoplasties (n = 454) during a 20-year period. At approximately the halfway point of this time frame, the abdominoplasty technique was changed from the use of 2 drains to the use of PTS without drains. Additionally, pulsed electromagnetic field therapy (PEMF) and liposomal bupivacaine (Exparel, Pacira Pharmaceuticals, Inc., Parsippany, NJ) were added as pain control adjuncts. RESULTS: There were 194 patients in the drain group and 260 patients in the PTS/no drains group. The group without drains contained a significantly higher proportion of massive weight loss patients (4.1% vs 9.2%, P = .041). The majority of the group without drains underwent outpatient surgery (89.7% vs 98.8%, P < .001). The overall complication rate was significantly lower in the no drains group (31.4% vs 13.8%, P < .001). The incidence of seroma was dramatically reduced in the group without drains (24.7% vs 0.0%, P < .001). CONCLUSIONS: PTS are highly effective in preventing seroma and can be safely employed as an alternative to drains in abdominoplasty. PEMF may play a role in seroma prevention and is also helpful for pain control. With these techniques to mitigate complications and minimize postoperative pain, abdominoplasty can be performed safely and effectively in a purely outpatient setting.

2.
Nature ; 477(7362): 99-102, 2011 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-21886163

RESUMO

The efficacy and safety of biological molecules in cancer therapy, such as peptides and small interfering RNAs (siRNAs), could be markedly increased if high concentrations could be achieved and amplified selectively in tumour tissues versus normal tissues after intravenous administration. This has not been achievable so far in humans. We hypothesized that a poxvirus, which evolved for blood-borne systemic spread in mammals, could be engineered for cancer-selective replication and used as a vehicle for the intravenous delivery and expression of transgenes in tumours. JX-594 is an oncolytic poxvirus engineered for replication, transgene expression and amplification in cancer cells harbouring activation of the epidermal growth factor receptor (EGFR)/Ras pathway, followed by cell lysis and anticancer immunity. Here we show in a clinical trial that JX-594 selectively infects, replicates and expresses transgene products in cancer tissue after intravenous infusion, in a dose-related fashion. Normal tissues were not affected clinically. This platform technology opens up the possibility of multifunctional products that selectively express high concentrations of several complementary therapeutic and imaging molecules in metastatic solid tumours in humans.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Poxviridae/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , DNA Viral/sangue , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Neoplasias/patologia , Neoplasias/cirurgia , Neoplasias/virologia , Organismos Geneticamente Modificados/fisiologia , Transgenes/genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
3.
Mol Ther ; 23(9): 1532-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26073886

RESUMO

Fifteen patients with treatment-refractory colorectal cancer were enrolled on a phase 1b study of Pexa-Vec (pexastimogene devacirepvec; JX-594), an oncolytic and immunotherapeutic vaccinia designed to selectively replicate in cancer cells. Pexa-Vec was administered intravenously every 14 days, at dose levels of 1 × 10(6), 1 × 10(7), or 3 × 10(7) plaque-forming units (pfu)/kg. The primary endpoint was to determine the maximum tolerated dose. Secondary endpoints were pharmacokinetics and pharmacodynamics as well as antitumor activity. Patients were heavily pretreated (mean 4.5 lines of therapy). All patients received at least two Pexa-Vec doses (median = 4; range = 2-4). No dose-limiting toxicities were reported, and the maximum tolerated dose was not reached. The most common adverse events were grade 1/2 flu-like symptoms, generally lasting <24 hours. During the first and last cycles, genome pharmacokinetics were unchanged. Infectious pfu could be detected in plasma up to 2 hours after cycle 1 and up to 30 minutes after cycle 4 (when antivaccinia antibody titers are known to have peaked). Ten patients (67%) had radiographically stable disease. Given the acceptable safety profile of multiple intravenous Pexa-Vec infusions in patients with treatment-refractory colorectal cancer, further trials evaluating efficacy of intravenous Pexa-Vec, as monotherapy or in combination with chemotherapeutic agents, is warranted in this patient population.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunoterapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vaccinia virus/genética , Administração Intravenosa , Adulto , Idoso , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Terapia Combinada , Citocinas/sangue , Esquema de Medicação , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Dermatopatias/etiologia , Dermatopatias/patologia , Resultado do Tratamento
4.
Mol Ther ; 23(3): 602-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25531693

RESUMO

Pexa-Vec (pexastimogene devacirepvec, JX-594) is an oncolytic and immunotherapeutic vaccinia virus designed to destroy cancer cells through viral lysis and induction of granulocyte-macrophage colony-stimulating factor (GM-CSF)-driven tumor-specific immunity. Pexa-Vec has undergone phase 1 and 2 testing alone and in combination with other therapies in adult patients, via both intratumoral and intravenous administration routes. We sought to determine the safety of intratumoral administration in pediatric patients. In a dose-escalation study using either 10(6) or 10(7) plaque-forming units per kilogram, we performed one-time injections in up to three tumor sites in five pediatric patients and two injections in one patient. Ages at study entry ranged from 4 to 21 years, and their cancer diagnoses included neuroblastoma, hepatocellular carcinoma, and Ewing sarcoma. All toxicities were ≤ grade 3. The most common side effects were sinus fever and sinus tachycardia. All three patients at the higher dose developed asymptomatic grade 1 treatment-related skin pustules that resolved within 3-4 weeks. One patient showed imaging evidence suggestive of antitumor biological activity. The two patients tested for cellular immunoreactivity to vaccinia antigens showed strong responses. Overall, our study suggests Pexa-Vec is safe to administer to pediatric patients by intratumoral administration and could be studied further in this patient population.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Vacinas Anticâncer/imunologia , Raios gama , Imunoterapia/métodos , Terapia Viral Oncolítica/métodos , Vaccinia virus/imunologia , Adolescente , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/administração & dosagem , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Criança , Pré-Escolar , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Injeções Intralesionais , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Masculino , Estadiamento de Neoplasias , Neuroblastoma/imunologia , Neuroblastoma/patologia , Neuroblastoma/terapia , Sarcoma de Ewing/imunologia , Sarcoma de Ewing/patologia , Sarcoma de Ewing/terapia , Vacinação , Vaccinia virus/genética , Adulto Jovem
5.
Mol Ther ; 23(1): 202-14, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25292189

RESUMO

Oncolytic viral therapy utilizes a tumor-selective replicating virus which preferentially infects and destroys cancer cells and triggers antitumor immunity. The Western Reserve strain of vaccinia virus (VV) is the most virulent strain of VV in animal models and has been engineered for tumor selectivity through two targeted gene deletions (vvDD). We performed the first-in-human phase 1, intratumoral dose escalation clinical trial of vvDD in 16 patients with advanced solid tumors. In addition to safety, we evaluated signs of vvDD replication and spread to distant tumors, pharmacokinetics and pharmacodynamics, clinical and immune responses to vvDD. Dose escalation proceeded without dose-limiting toxicities to a maximum feasible dose of 3 × 10(9) pfu. vvDD replication in tumors was reproducible. vvDD genomes and/or infectious particles were recovered from injected (n = 5 patients) and noninjected (n = 2 patients) tumors. At the two highest doses, vvDD genomes were detected acutely in blood in all patients while delayed re-emergence of vvDD genomes in blood was detected in two patients. Fifteen of 16 patients exhibited late symptoms, consistent with ongoing vvDD replication. In summary, intratumoral injection of the oncolytic vaccinia vvDD was well-tolerated in patients and resulted in selective infection of injected and noninjected tumors and antitumor activity.


Assuntos
Neoplasias da Mama/terapia , Neoplasias do Colo/terapia , Melanoma/terapia , Neoplasias Pancreáticas/terapia , Neoplasias Cutâneas/terapia , Vaccinia virus/imunologia , Replicação Viral/genética , Idoso , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Relação Dose-Resposta Imunológica , Feminino , Deleção de Genes , Humanos , Injeções Intralesionais , Masculino , Melanoma/imunologia , Melanoma/patologia , Pessoa de Meia-Idade , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/imunologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Vaccinia virus/genética , Vaccinia virus/crescimento & desenvolvimento
6.
Mol Ther ; 23(6): 1066-1076, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807289

RESUMO

Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.


Assuntos
Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Vaccinia virus/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Estudos de Viabilidade , Feminino , Células HeLa , Humanos , Injeções Intralesionais , Macaca fascicularis/imunologia , Masculino , Neoplasias/sangue , Neoplasias/terapia , Testes de Neutralização , Piridonas/imunologia , Piridonas/farmacologia , Ratos , Ratos Endogâmicos F344 , Vacina Antivariólica/sangue , Vacina Antivariólica/imunologia , Vacinação , Células Vero
7.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37988172

RESUMO

BACKGROUNDSystemic administration of adeno-associated virus (AAV) can trigger life-threatening inflammatory responses, including thrombotic microangiopathy (TMA), acute kidney injury due to atypical hemolytic uremic syndrome-like complement activation, immune-mediated myocardial inflammation, and hepatic toxicity.METHODSWe describe the kinetics of immune activation following systemic AAV serotype 9 (AAV9) administration in 38 individuals following 2 distinct prophylactic immunomodulation regimens. Group 1 received corticosteroids and Group 2 received rituximab plus sirolimus in addition to steroids to prevent anti-AAV antibody formation.RESULTSGroup 1 participants had a rapid increase in immunoglobulin M (IgM) and IgG. Increase in D-dimer, decline in platelet count, and complement activation are indicative of TMA. All Group 1 participants demonstrated activation of both classical and alternative complement pathways, as indicated by depleted C4 and elevated soluble C5b-9, Ba, and Bb antigens. Group 2 patients did not have a significant change in IgM or IgG and had minimal complement activation.CONCLUSIONSThis study demonstrates that TMA in the setting of AAV gene therapy is antibody dependent (classical pathway) and amplified by the alternative complement pathway. Critical time points and interventions are identified to allow for management of immune-mediated events that impact the safety and efficacy of systemic gene therapy.


Assuntos
Dependovirus , Microangiopatias Trombóticas , Humanos , Dependovirus/genética , Microangiopatias Trombóticas/terapia , Imunoglobulina M , Imunoglobulina G
8.
Mol Ther ; 20(4): 749-58, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22186794

RESUMO

Oncolytic viruses are generally designed to be cancer selective on the basis of a single genetic mutation. JX-594 is a thymidine kinase (TK) gene-inactivated oncolytic vaccinia virus expressing granulocyte-macrophage colony-stimulating factor (GM-CSF) and lac-Z transgenes that is designed to destroy cancer cells through replication-dependent cell lysis and stimulation of antitumoral immunity. JX-594 has demonstrated a favorable safety profile and reproducible tumor necrosis in a variety of solid cancer types in clinical trials. However, the mechanism(s) responsible for its cancer-selectivity have not yet been well described. We analyzed the replication of JX-594 in three model systems: primary normal and cancer cells, surgical explants, and murine tumor models. JX-594 replication, transgene expression, and cytopathic effects were highly cancer-selective, and broad spectrum activity was demonstrated. JX-594 cancer-selectivity was multi-mechanistic; replication was activated by epidermal growth factor receptor (EGFR)/Ras pathway signaling, cellular TK levels, and cancer cell resistance to type-I interferons (IFNs). These findings confirm a large therapeutic index for JX-594 that is driven by common genetic abnormalities in human solid tumors. This appears to be the first description of multiple selectivity mechanisms, both inherent and engineered, for an oncolytic virus. These findings have implications for oncolytic viruses in general, and suggest that their cancer targeting is a complex and multifactorial process.


Assuntos
Neoplasias/metabolismo , Vírus Oncolíticos/fisiologia , Poxviridae/fisiologia , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HeLa , Humanos , Técnicas In Vitro , Leucócitos Mononucleares , Camundongos , Camundongos Nus , Neoplasias/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Poxviridae/genética , Transdução de Sinais/genética , Replicação Viral/genética
9.
Mol Ther ; 19(10): 1913-22, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21772252

RESUMO

JX-594 is a targeted and granulocyte macrophage-colony stimulating factor (GM-CSF)-expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In order to study the mechanisms-of-action (MOA) of JX-594 in humans, a mechanistic proof-of-concept clinical trial was performed at a low dose equivalent to ≤10% of the maximum-tolerated dose (MTD) in other clinical trials. Ten patients with previously treated stage IV melanoma were enrolled. Tumors were injected weekly for up to nine total treatments. Blood samples and tumor biopsies were analyzed for evidence of transgene activity, virus replication, and immune stimulation. The ß-galactosidase (ß-gal) transgene was expressed in all patients as evidenced by antibody induction. Six patients had significant induction of GM-CSF-responsive white blood cell (WBC) subsets such as neutrophils (25-300% increase). JX-594 replication and subsequent shedding into blood was detectable in five patients after cycles 1-9. Tumor biopsies demonstrated JX-594 replication, perivascular lymphocytic infiltration, and diffuse tumor necrosis. Mild flu-like symptoms were the most common adverse events. In sum, JX-594 replication, oncolysis, and expression of both transgenes were demonstrated; replication was still evident after multiple cycles. These findings have implications for further clinical development of JX-594 and other transgene-armed oncolytic viruses.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Melanoma/terapia , Terapia Viral Oncolítica , Poxviridae/genética , Adulto , Idoso , Feminino , Humanos , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Poxviridae/fisiologia , Transgenes
10.
Mol Ther ; 19(6): 1170-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21427706

RESUMO

JX-594 is a targeted and granulocyte-macrophage colony stimulating factor (GM-CSF) expressing oncolytic poxvirus designed to selectively replicate in and destroy cancer cells through viral oncolysis and tumor-specific immunity. In a phase 1 trial, JX-594 injection into hepatocellular carcinoma (HCC) was well-tolerated and associated with viral replication, decreased tumor perfusion, and tumor necrosis. We hypothesized that JX-594 and sorafenib, a small molecule inhibitor of B-raf and vascular endothelial growth factor receptor (VEGFR) approved for HCC, would have clinical benefit in combination given their demonstrated efficacy in HCC patients and their complementary mechanisms-of-action. HCC cell lines were uniformly sensitive to JX-594. Anti-raf kinase effects of concurrent sorafenib inhibited JX-594 replication in vitro, whereas sequential therapy was superior to either agent alone in murine tumor models. We therefore explored pilot safety and efficacy of JX-594 followed by sorafenib in three HCC patients. In all three patients, sequential treatment was (i) well-tolerated, (ii) associated with significantly decreased tumor perfusion, and (iii) associated with objective tumor responses (Choi criteria; up to 100% necrosis). HCC historical control patients on sorafenib alone at the same institutions had no objective tumor responses (0 of 15). Treatment of HCC with JX-594 followed by sorafenib has antitumoral activity, and JX-594 may sensitize tumors to subsequent therapy with VEGF/VEGFR inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Benzenossulfonatos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/terapia , Piridinas/uso terapêutico , Vaccinia virus/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/terapia , Melanoma/tratamento farmacológico , Melanoma/terapia , Camundongos , Camundongos SCID , Niacinamida/análogos & derivados , Terapia Viral Oncolítica/métodos , Compostos de Fenilureia , Sorafenibe , Vaccinia virus/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Mol Ther ; 19(5): 886-94, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21364541

RESUMO

Oncolytic viruses (OVs) have been engineered or selected for cancer cell-specific infection however, we have found that following intravenous administration of vesicular stomatitis virus (VSV), tumor cell killing rapidly extends far beyond the initial sites of infection. We show here for the first time that VSV directly infects and destroys tumor vasculature in vivo but leaves normal vasculature intact. Three-dimensional (3D) reconstruction of infected tumors revealed that the majority of the tumor mass lacks significant blood flow in contrast to uninfected tumors, which exhibit relatively uniform perfusion. VSV replication in tumor neovasculature and spread within the tumor mass, initiates an inflammatory reaction including a neutrophil-dependent initiation of microclots within tumor blood vessels. Within 6 hours of intravenous administration of VSV and continuing for at least 24 hours, we observed the initiation of blood clots within the tumor vasculature whereas normal vasculature remained clot free. Blocking blood clot formation with thrombin inhibitors prevented tumor vascular collapse. Our results demonstrate that the therapeutic activity of an OV can go far beyond simple infection and lysis of malignant cells.


Assuntos
Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus da Estomatite Vesicular Indiana , Adenocarcinoma/genética , Animais , Coagulação Sanguínea , Linhagem Celular Tumoral , Proliferação de Células , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos , Trombina/antagonistas & inibidores
12.
Mol Ther ; 18(5): 888-95, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20234341

RESUMO

A major barrier to all oncolytic viruses (OVs) in clinical development is cellular innate immunity, which is variably active in a spectrum of human malignancies. To overcome the heterogeneity of tumor response, we combined complementary OVs that attack cancers in distinct ways to improve therapeutic outcome. Two genetically distinct viruses, vesicular stomatitis virus (VSV) and vaccinia virus (VV), were used to eliminate the risk of recombination. The combination was tested in a variety of tumor types in vitro, in immunodeficient and immunocompetent mouse tumor models, and ex vivo, in a panel of primary human cancer samples. We found that VV synergistically enhanced VSV antitumor activity, dependent in large part on the activity of the VV B18R gene product. A recombinant version of VSV expressing the fusion-associated small-transmembrane (p14FAST) protein also further enhanced the ability of VV to spread through an infected monolayer, resulting in a "ping pong" oncolytic effect wherein each virus enhanced the ability of the other to replicate and/or spread in tumor cells. Our strategy is the first example where OVs are rationally combined to utilize attributes of different OVs to overcome the heterogeneity of malignancies and demonstrates the feasibility of combining complementary OVs to improve therapeutic outcome.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica/efeitos adversos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Chlorocebus aethiops , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/genética , Células HT29 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Vaccinia virus/genética , Vaccinia virus/fisiologia , Células Vero , Vesiculovirus/genética , Vesiculovirus/fisiologia
13.
Mol Ther ; 18(11): 1927-36, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20808290

RESUMO

The purpose of this study was to investigate the oncolytic potential of the recombinant, granulocyte macrophage colony-stimulating factor (GM-CSF)-expressing vaccinia virus (VV) JX-594 in experimental malignant glioma (MGs) in vitro and in immunocompetent rodent models. We have found that JX-594 killed all MG cell lines tested in vitro. Intratumoral (i.t.) administration of JX-594 significantly inhibited tumor growth and prolonged survival in rats-bearing RG2 intracranial (i.c.) tumors and mice-bearing GL261 brain tumors. Combination therapy with JX-594 and rapamycin significantly increased viral replication and further prolonged survival in both immunocompetent i.c. MG models with several animals considered "cured" (three out of seven rats >120 days, terminated experiment). JX-594 infected and killed brain tumor-initiating cells (BTICs) from patient samples grown ex vivo, and did so more efficiently than other oncolytic viruses MYXV, Reovirus type-3, and VSV(ΔM51). Additional safety/toxicity studies in nontumor-bearing rodents treated with a supratherapeutic dose of JX-594 demonstrated GM-CSF-dependent inflammation and necrosis. These results suggest that i.c. administered JX-594 triggers a predictable GM-CSF-mediated inflammation in murine models. Before proceeding to clinical trials, JX-594 should be evaluated in the brains of nonhuman primates and optimized for the viral doses, delivery routes as well as the combination agents (e.g., mTOR inhibitors).


Assuntos
Neoplasias Encefálicas/terapia , Modelos Animais de Doenças , Glioma/terapia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Terapia Viral Oncolítica , Sirolimo/uso terapêutico , Vaccinia virus/genética , Animais , Antibióticos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/genética , Terapia Combinada , Feminino , Vetores Genéticos/uso terapêutico , Glioma/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/análise , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida , Transgenes/fisiologia , Células Tumorais Cultivadas , Vacinas Sintéticas/uso terapêutico , Replicação Viral
14.
Mol Ther ; 18(6): 1123-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20389287

RESUMO

Oncolytic viruses (OVs) are promising anticancer agents but like other cancer monotherapies, the genetic heterogeneity of human malignancies can lead to treatment resistance. We used a virus/cell-based assay to screen diverse chemical libraries to identify small molecules that could act in synergy with OVs to destroy tumor cells that resist viral infection. Several molecules were identified that aid in viral oncolysis, enhancing virus replication and spread as much as 1,000-fold in tumor cells. One of these molecules we named virus-sensitizers 1 (VSe1), was found to target tumor innate immune response and could enhance OV efficacy in animal tumor models and within primary human tumor explants while remaining benign to normal tissues. We believe this is the first example of a virus/cell-based "pharmacoviral" screen aimed to identify small molecules that modulate cellular response to virus infection and enhance oncolytic virotherapy.


Assuntos
Terapia Viral Oncolítica , Animais , Linhagem Celular Tumoral , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia
15.
Proc Natl Acad Sci U S A ; 105(39): 14981-6, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18815361

RESUMO

Intratumoral innate immunity can play a significant role in blocking the effective therapeutic spread of a number of oncolytic viruses (OVs). Histone deacetylase inhibitors (HDIs) are known to influence epigenetic modifications of chromatin and can blunt the cellular antiviral response. We reasoned that pretreatment of tumors with HDIs could enhance the replication and spread of OVs within malignancies. Here, we show that HDIs markedly enhance the spread of vesicular stomatitis virus (VSV) in a variety of cancer cells in vitro, in primary tumor tissue explants and in multiple animal models. This increased oncolytic activity correlated with a dampening of cellular IFN responses and augmentation of virus-induced apoptosis. These results illustrate the general utility of HDIs as chemical switches to regulate cellular innate antiviral responses and to provide controlled growth of therapeutic viruses within malignancies. HDIs could have a profoundly positive impact on the clinical implementation of OV therapeutics.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Inibidores de Histona Desacetilases , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/efeitos dos fármacos , Animais , Benzamidas/uso terapêutico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Interferons/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos , Neoplasias/tratamento farmacológico , Neoplasias/virologia , Vírus Oncolíticos/imunologia , Vírus Oncolíticos/fisiologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/terapia , Neoplasias da Próstata/virologia , Piridinas/uso terapêutico , Vesiculovirus/efeitos dos fármacos , Vesiculovirus/imunologia , Vesiculovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Mol Cancer Ther ; 20(8): 1481-1494, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34045231

RESUMO

Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.


Assuntos
Apoptose , Citocinas/metabolismo , Imunidade , Tumores Neuroendócrinos/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Vaccinia virus/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-2/genética , Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/virologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/virologia , Células Tumorais Cultivadas
17.
J Clin Invest ; 117(11): 3350-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17965776

RESUMO

Replication-selective oncolytic viruses (virotherapeutics) are being developed as novel cancer therapies with unique mechanisms of action, but limitations in i.v. delivery to tumors and systemic efficacy have highlighted the need for improved agents for this therapeutic class to realize its potential. Here we describe the rational, stepwise design and evaluation of a systemically effective virotherapeutic (JX-963). We first identified a highly potent poxvirus strain that also trafficked efficiently to human tumors after i.v. administration. This strain was then engineered to target cancer cells with activation of the transcription factor E2F and the EGFR pathway by deletion of the thymidine kinase and vaccinia growth factor genes. For induction of tumor-specific cytotoxic T lymphocytes, we further engineered the virus to express human GM-CSF. JX-963 was more potent than the previously used virotherapeutic Onyx-015 adenovirus and as potent as wild-type vaccinia in all cancer cell lines tested. Significant cancer selectivity of JX-963 was demonstrated in vitro in human tumor cell lines, in vivo in tumor-bearing rabbits, and in primary human surgical samples ex vivo. Intravenous administration led to systemic efficacy against both primary carcinomas and widespread organ-based metastases in immunocompetent mice and rabbits. JX-963 therefore holds promise as a rationally designed, targeted virotherapeutic for the systemic treatment of cancer in humans and warrants clinical testing.


Assuntos
Neoplasias/metabolismo , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Poxviridae/fisiologia , Animais , Linhagem Celular Tumoral , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/genética , Vírus Oncolíticos/genética , Poxviridae/genética , Coelhos
18.
Transl Vis Sci Technol ; 9(7): 21, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32832227

RESUMO

Purpose: To develop a reliable and efficient method for quantifying the area of preserved retinal pigment epithelium (RPE), facilitating the evaluation of disease progression or response to therapy in choroideremia (CHM). Methods: The fundus autofluorescence images of CHM patients were captured at baseline and 1 year. A Photoshop-based method was developed to allow the reliable measurement of the RPE area. The results were compared with measurements generated by the Heidelberg Eye Explorer 2 (HEYEX2). The areas measured by two independent graders were compared to assess the test-retest reliability. Results: By using the Photoshop-based method, the area of the RPE measured from 64 eyes was seen to decrease significantly (P < 0.001) at a rate of 2.57 ± 3.22 mm2 annually, and a percentage of 8.39% ± 5.24%. The average standard deviations for Photoshop were less than that for HEYEX2 (0.5-1.1 in grader 1; 0.4-1.6 in grader 2), indicating less intragrader variability. The RPE decrease as determined by the Photoshop-based method showed excellent reliability with an intraclass correlation coefficient of 0.944 (95% confidence interval, 0.907-0.966). In Bland-Altman plots, the Photoshop method also exhibited better intergrader agreement. Conclusions: Photoshop-based quantification of preserved RPE area in patients with CHM is feasible and has better test-retest reliability compared with the HEYEX2 method. Translational Relevance: An accurate quantification method for longitudinal RPE change in CHM patients is an important tool for the evaluation of efficacy in any therapeutic trials.


Assuntos
Coroideremia , Humanos , Reprodutibilidade dos Testes , Epitélio Pigmentado da Retina , Tomografia de Coerência Óptica , Acuidade Visual
19.
J Cell Mol Med ; 13(7): 1238-47, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19175689

RESUMO

Primary and metastatic neoplasms of the liver account for more than a million deaths per year worldwide. Despite decades of research, effective novel therapies for these cancers are urgently needed. Oncolytic virotherapeutics represent a novel class of pharmacophore that holds promise for the treatment of hepatic neoplasms. Cancer-specific replication is followed by oncolysis, virus spreading and infection of adjacent cancer cells. This process is then repeated. Virotherapeutics target multiple genetic pathways involved in carcino-genesis, and demonstrate activity against apoptosis-resistant tumour cells. This platform can also exploit the advantage of multiple intrinsic anti-cancer therapeutic mechanisms, combining direct viral oncolysis with therapeutic transgene expression. Recent advances in pre-clinical and clinical studies are revealing the potential of this unique therapeutic class, in particular for liver cancers. This review summarizes the available data on applying oncolytic virotherapeutics to hepatic neoplasms to date, and discusses the challenges and future directions for virotherapy.


Assuntos
Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica/métodos , Animais , Ensaios Clínicos como Assunto , Ensaios de Seleção de Medicamentos Antitumorais , Humanos
20.
Lung Cancer ; 63(1): 151-3, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18571759

RESUMO

A 46-year-old nonsmoking female was diagnosed with pulmonary lymphoepithelioma-like carcinoma (LELC). She simultaneously had a rare skin manifestation, erythema elevatum diutinum (EED), which did not respond to topical treatment. The patient underwent platinum-based chemotherapy and thoracic radiotherapy and had shown complete remission in both LELC and EED. EED is therefore considered as a paraneoplastic syndrome of pulmonary LELC in this case. Literature on LELC and EED were also reviewed.


Assuntos
Carcinoma/diagnóstico , Eritema/diagnóstico , Neoplasias Pulmonares/diagnóstico , Síndromes Paraneoplásicas/diagnóstico , Antineoplásicos/farmacologia , Biópsia , Carcinoma/complicações , Eritema/complicações , Feminino , Humanos , Pulmão/patologia , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/patologia , Pessoa de Meia-Idade , Platina/farmacologia , Indução de Remissão , Pele/patologia , Tomografia Computadorizada por Raios X/métodos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA