Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Mol Sci ; 21(14)2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32708234

RESUMO

Oncolytic adenoviruses (OAds) present limited efficacy in clinics. The insertion of therapeutic transgenes into OAds genomes, known as "arming OAds", has been the main strategy to improve their therapeutic potential. Different approaches were published in the decade of the 2000s, but with few comparisons. Most armed OAds have complete or partial E3 deletions, leading to a shorter half-life in vivo. We generated E3+ OAds using two insertion sites, After-fiber and After-E4, and two different splice acceptors linked to the major late promoter, either the Ad5 protein IIIa acceptor (IIIaSA) or the Ad40 long fiber acceptor (40SA). The highest transgene levels were obtained with the After-fiber location and 40SA. However, the set of codons of the transgene affected viral fitness, highlighting the relevance of transgene codon usage when arming OAds using the major late promoter.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vírus Oncolíticos/genética , Replicação Viral/genética , Adenoviridae/metabolismo , Animais , Linhagem Celular Tumoral , Uso do Códon , Genes Reporter , Vetores Genéticos , Humanos , Camundongos , Vírus Oncolíticos/metabolismo , Análise de Componente Principal , Regiões Promotoras Genéticas , Transgenes , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Hyperthermia ; 29(2): 133-44, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23406389

RESUMO

PURPOSE: The present study aims to create and characterise a cell-embedding tissue-mimicking material (TMM) that has thermal and acoustic properties similar to liver tissue, in order to enable study and optimisation of protocols for ultrasound-induced hyperthermia and drug delivery. MATERIALS AND METHODS: An agarose-based, cell-embedding TMM was iteratively developed and characterised. The acoustic properties (attenuation coefficient, speed of sound and cavitation threshold) and thermal response of the material were compared with those of fresh degassed liver tissue over a range of acoustic pressures and frequencies. A luminescence intensity assay was used to evaluate viability of HuH-7 cells in the material. The efficacy of ultrasound-mediated chemotherapeutic treatment in the material was tested by localised activation of low temperature thermally sensitive liposomes. Drug activation was measured by fluorescence microscopy. RESULTS: Similar acoustic properties (attenuation coefficient, speed of sound) to liver tissue were achieved over the therapeutically relevant frequency range of 1-4 MHz and similar thermal response was achieved for acoustic pressures up to 4.8 MPa peak to peak (ppk) at 1.1 MHz. Above 4.8 MPa ppk cavitation enhanced heating occurred in the TMM. Drug release from low-temperature-sensitive liposomes was achieved with 4.4 MPa ppk 6-s exposures at 1.1 MHz and cell compatibility of the material was confirmed. CONCLUSIONS: A platform for in vitro work for activation of thermally sensitive liposomes using high intensity focused ultrasound (HIFU)-induced hyperthermia was established. The TMM presents similar acoustic properties and thermal response to liver tissue over a broad range of ultrasound exposure conditions.


Assuntos
Sistemas de Liberação de Medicamentos , Ablação por Ultrassom Focalizado de Alta Intensidade/métodos , Fígado , Inclusão do Tecido , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Sobrevivência Celular , Doxorrubicina/administração & dosagem , Géis , Humanos , Lipossomos , Neoplasias Hepáticas/terapia , Imagens de Fantasmas , Sefarose
3.
Circ Res ; 107(2): 283-93, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20508179

RESUMO

RATIONALE: Notch signaling regulates vascular development. However, the implication of the Notch ligand Delta-like 4 (Dll4) in postischemic angiogenesis remains unclear. OBJECTIVE: We investigated the role of Dll4/Notch signaling in reparative angiogenesis using a mouse model of ischemia. METHODS AND RESULTS: We found Dll4 weakly expressed in microvascular endothelial cells of normoperfused muscles. Conversely, Dll4 is upregulated following ischemia and localized at the forefront of sprouting capillaries. We analyzed the effect of inhibiting endogenous Dll4 by intramuscular injection of an adenovirus encoding the soluble form of Dll4 extracellular domain (Ad-sDll4). Dll4 inhibition caused the formation of a disorganized, low-perfused capillary network in ischemic muscles. This structural abnormality was associated to delayed blood flow recovery and muscle hypoxia and degeneration. Analysis of microvasculature at early stages of repair revealed that Dll4 inhibition enhances capillary sprouting in a chaotic fashion and causes excessive leukocyte infiltration of ischemic muscles. Furthermore, Dll4 inhibition potentiated the elevation of the leukocyte chemoattractant CXCL1 (chemokine [C-X-C motif] ligand 1) following ischemia, without altering peripheral blood levels of stromal cell-derived factor-1 and monocyte chemoattractant protein-1. In cultured human monocytes, Dll4 induces the transcription of Notch target gene Hes-1 and inhibits the basal and tumor necrosis factor-alpha-stimulated production of interleukin-8, the human functional homolog of murine CXCL1. The inhibitory effect of Dll4 on interleukin-8 was abolished by DAPT, a Notch inhibitor, or by coculturing activated human monocytes with Ad-sDll4-infected endothelial cells. CONCLUSIONS: Dll4/Notch interaction is essential for proper reparative angiogenesis. Moreover, Dll4/Notch signaling regulates sprouting angiogenesis and coordinates the interaction between inflammation and angiogenesis under ischemic conditions.


Assuntos
Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Receptores Notch/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Quimiocina CXCL1/metabolismo , Quimiotaxia de Leucócito , Técnicas de Cocultura , Modelos Animais de Doenças , Membro Posterior , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interleucina-8/metabolismo , Isquemia/diagnóstico por imagem , Isquemia/genética , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Leucócitos/metabolismo , Masculino , Camundongos , Neovascularização Fisiológica/genética , Regeneração , Fluxo Sanguíneo Regional , Transdução de Sinais/genética , Fatores de Tempo , Transfecção , Ultrassonografia
4.
Mol Ther ; 19(1): 67-75, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20877345

RESUMO

The endothelium imposes a structural barrier to the extravasation of systemically delivered oncolytic adenovirus (Ad). Here, we introduced a transendothelial route of delivery in order to increase tumor accumulation of virus particles (vp) beyond that resulting from convection-dependent extravasation alone. This was achieved by engineering an Ad encoding a syncytium-forming protein, gibbon ape leukemia virus (GALV) fusogenic membrane glycoprotein (FMG). The expression of GALV was regulated by a hybrid viral enhancer-human promoter construct comprising the human cytomegalovirus (CMV) immediate-early enhancer and the minimal human endothelial receptor tyrosine kinase promoter ("eTie1"). Endothelial cell-selectivity of the resulting Ad-eTie1-GALV vector was demonstrated by measuring GALV mRNA transcript levels. Furthermore, Ad-eTie1-GALV selectively induced fusion between infected endothelial cells and uninfected epithelial cells in vitro and in vivo, allowing transendothelial virus penetration. Heterofusion of infected endothelium to human embryonic kidney 293 (HEK 293) cells, in mixed in vitro cultures or in murine xenograft models, permitted fusion-dependent transactivation of the replication-deficient Ad-eTie1-GALV, due to enabled access to viral E1 proteins derived from the HEK 293 cytoplasm. These data provide evidence to support our proposed use of GALV to promote Ad penetration through tumor-associated vasculature, an approach that may substantially improve the efficiency of systemic delivery of oncolytic viruses to disseminated tumors.


Assuntos
Adenoviridae/metabolismo , Fusão Celular/métodos , Células Gigantes/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Glicoproteínas de Membrana/genética , Terapia de Alvo Molecular/métodos , Migração Transendotelial e Transepitelial/genética , Adenoviridae/genética , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/virologia , Elementos Facilitadores Genéticos , Células Epiteliais/metabolismo , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células Gigantes/citologia , Células HEK293 , Hepatócitos/citologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos SCID , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica/métodos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transplante Heterólogo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/farmacologia , Vírion , Replicação Viral/genética
5.
J Immunother Cancer ; 10(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35338084

RESUMO

BACKGROUND: VCN-01 is an oncolytic adenovirus (Ad5 based) designed to replicate in cancer cells with dysfunctional RB1 pathway, express hyaluronidase to enhance virus intratumoral spread and facilitate chemotherapy and immune cells extravasation into the tumor. This phase I clinical trial was aimed to find the maximum tolerated dose/recommended phase II dose (RP2D) and dose-limiting toxicity (DLT) of the intravenous delivery of the replication-competent VCN-01 adenovirus in patients with advanced cancer. METHODS: Part I: patients with advanced refractory solid tumors received one single dose of VCN-01. Parts II and III: patients with pancreatic adenocarcinoma received VCN-01 (only in cycle 1) and nab-paclitaxel plus gemcitabine (VCN-concurrent on day 1 in Part II, and 7 days before chemotherapy in Part III). Patients were required to have anti-Ad5 neutralizing antibody (NAbs) titers lower than 1/350 dilution. Pharmacokinetic and pharmacodynamic analyses were performed. RESULTS: 26% of the patients initially screened were excluded based on high NAbs levels. Sixteen and 12 patients were enrolled in Part I and II, respectively: RP2D were 1×1013 viral particles (vp)/patient (Part I), and 3.3×1012 vp/patient (Part II). Fourteen patients were included in Part III: there were no DLTs and the RP2D was 1×1013 vp/patient. Observed DLTs were grade 4 aspartate aminotransferase increase in one patient (Part I, 1×1013 vp), grade 4 febrile neutropenia in one patient and grade 5 thrombocytopenia plus enterocolitis in another patient (Part II, 1×1013 vp). In patients with pancreatic adenocarcinoma overall response rate were 50% (Part II) and 50% (Part III). VCN-01 viral genomes were detected in tumor tissue in five out of six biopsies (day 8). A second viral plasmatic peak and increased hyaluronidase serum levels suggested replication after intravenous injection in all patients. Increased levels of immune biomarkers (interferon-γ, soluble lymphocyte activation gene-3, interleukin (IL)-6, IL-10) were found after VCN-01 administration. CONCLUSIONS: Treatment with VCN-01 is feasible and has an acceptable safety. Encouraging biological and clinical activity was observed when administered in combination with nab-paclitaxel plus gemcitabine to patients with pancreatic adenocarcinoma. TRIAL REGISTRATION NUMBER: NCT02045602.


Assuntos
Adenocarcinoma , Neoplasias Pancreáticas , Adenocarcinoma/patologia , Adenoviridae/genética , Albuminas , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Desoxicitidina/análogos & derivados , Humanos , Hialuronoglucosaminidase/uso terapêutico , Paclitaxel , Neoplasias Pancreáticas/tratamento farmacológico , Gencitabina , Neoplasias Pancreáticas
6.
PLoS Pathog ; 5(5): e1000440, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19461878

RESUMO

Replicating viruses have broad applications in biomedicine, notably in cancer virotherapy and in the design of attenuated vaccines; however, uncontrolled virus replication in vulnerable tissues can give pathology and often restricts the use of potent strains. Increased knowledge of tissue-selective microRNA expression now affords the possibility of engineering replicating viruses that are attenuated at the RNA level in sites of potential pathology, but retain wild-type replication activity at sites not expressing the relevant microRNA. To assess the usefulness of this approach for the DNA virus adenovirus, we have engineered a hepatocyte-safe wild-type adenovirus 5 (Ad5), which normally mediates significant toxicity and is potentially lethal in mice. To do this, we have included binding sites for hepatocyte-selective microRNA mir-122 within the 3' UTR of the E1A transcription cassette. Imaging versions of these viruses, produced by fusing E1A with luciferase, showed that inclusion of mir-122 binding sites caused up to 80-fold decreased hepatic expression of E1A following intravenous delivery to mice. Animals administered a ten-times lethal dose of wild-type Ad5 (5x10(10) viral particles/mouse) showed substantial hepatic genome replication and extensive liver pathology, while inclusion of 4 microRNA binding sites decreased replication 50-fold and virtually abrogated liver toxicity. This modified wild-type virus retained full activity within cancer cells and provided a potent, liver-safe oncolytic virus. In addition to providing many potent new viruses for cancer virotherapy, microRNA control of virus replication should provide a new strategy for designing safe attenuated vaccines applied across a broad range of viral diseases.


Assuntos
Adenoviridae/fisiologia , Hepatócitos/virologia , MicroRNAs/metabolismo , Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Alanina Transaminase/sangue , Sequência de Aminoácidos , Animais , Fusão Gênica Artificial , Aspartato Aminotransferases/sangue , Sítios de Ligação/genética , Linhagem Celular Tumoral , Fluorescência , Regulação Viral da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/virologia , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Dados de Sequência Molecular , Terapia Viral Oncolítica , Distribuição Tecidual , Imagem Corporal Total
7.
Clin Cancer Res ; 27(11): 2979-2988, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33526422

RESUMO

Cancer immunotherapy targeting immune checkpoint inhibitors shows efficacy in several human cancers, but "cold tumors" that lack immune cells are typically unresponsive. Among the potential therapeutic approaches that could "heat" or promote lymphocyte infiltration of cold tumors, oncolytic viruses have attracted interest for their lytic and immunogenic mechanisms of action. In this article, we review the use of oncolytic adenoviruses in cancer immunotherapy, with a particular focus on preclinical and clinical data of oncolytic adenovirus-triggered immune responses against tumor antigens. We also discuss parameters to consider in clinical trial design and the combination of oncolytic adenoviruses with conventional treatments or other immunotherapies.


Assuntos
Adenoviridae/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Antígenos de Neoplasias/imunologia , Humanos , Tolerância Imunológica/imunologia , Imunoterapia/tendências , Terapia Viral Oncolítica/tendências , Linfócitos T/imunologia
8.
Mol Ther Oncolytics ; 22: 27-35, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34377767

RESUMO

Oncolytic viruses (OVs) preferentially infect and selectively replicate in cancer cells. OVs have been tested in clinical trials as monotherapy or in combination with chemotherapy, radiotherapy, and immunotherapy. However, the dense extracellular matrix hampers the intratumoral spreading and efficacy of OVs. Previously we described VCN-01, an oncolytic adenovirus expressing a soluble version of human sperm hyaluronidase (hyal) PH20, which exhibited enhanced intratumoral distribution and antitumor activity in different models. Here, we present two oncolytic adenoviruses designed to increase the secretion of PH20 compared to VCN-01. ICO15K-40SAPH20, encoding PH20 under an Ad40 splice acceptor, and ICO15K-E1aPH20 expressing PH20 fused to the E1A gene by P2A peptide. We demonstrate that increased hyal activity improves antitumor efficacy in both a sensitive immunodeficient model and an immunocompetent model. Moreover, we show that hyal activity impacts T cell accumulation in tumors, highlighting the value of a hyaluronidase-expressing virus for combinations with other immunotherapies in cancers involving dense stroma.

9.
J Control Release ; 332: 517-528, 2021 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-33675877

RESUMO

Tumor targeting and intratumoral virus spreading are key features for successful oncolytic virotherapy. VCN-11 is a novel oncolytic adenovirus, genetically modified to express hyaluronidase (PH20) and display an albumin-binding domain (ABD) on the hexon. ABD allows the virus to self-coat with albumin when entering the bloodstream and evade neutralizing antibodies (NAbs). Here, we validate VCN-11 mechanism of action and characterize its toxicity. VCN-11 replication, hyaluronidase activity and binding to human albumin to evade NAbs was evaluated. Toxicity and efficacy of VCN-11 were assessed in mice and hamsters. Tumor targeting, and antitumor activity was analyzed in the presence of NAbs in several tumor models. VCN-11 induced 450 times more cytotoxicity in tumor cells than in normal cells. VCN-11 hyaluronidase production was confirmed by measuring PH20 activity in vitro and in virus-infected tumor areas in vivo. VCN-11 evaded NAbs from different sources and tumor targeting was demonstrated in the presence of high levels of NAbs in vivo, whereas the control virus without ABD was neutralized. VCN-11 showed a low toxicity profile in athymic nude mice and Syrian hamsters, allowing treatments with high doses and fractionated administrations without major toxicities (up to 1.2x1011vp/mouse and 7.5x1011vp/hamster). Fractionated intravenous administrations improved circulation kinetics and tumor targeting. VCN-11 antitumor efficacy was demonstrated in the presence of NAbs against Ad5 and itself. Oncolytic adenovirus VCN-11 disrupts tumor matrix and displays antitumor effects even in the presence of NAbs. These features make VCN-11 a safe promising candidate to test re-administration in clinical trials.


Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae , Animais , Anticorpos Neutralizantes , Linhagem Celular Tumoral , Cricetinae , Hialuronoglucosaminidase , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Immunother Cancer ; 9(11)2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149591

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is characterized by dense desmoplastic stroma that limits the delivery of anticancer agents. VCN-01 is an oncolytic adenovirus designed to replicate in cancer cells with a dysfunctional RB1 pathway and express hyaluronidase. Here, we evaluated the mechanism of action of VCN-01 in preclinical models and in patients with pancreatic cancer. METHODS: VCN-01 replication and antitumor efficacy were evaluated alone and in combination with standard chemotherapy in immunodeficient and immunocompetent preclinical models using intravenous or intratumoral administration. Hyaluronidase activity was evaluated by histochemical staining and by measuring drug delivery into tumors. In a proof-of-concept clinical trial, VCN-01 was administered intratumorally to patients with PDAC at doses up to 1×1011 viral particles in combination with chemotherapy. Hyaluronidase expression was measured in serum by an ELISA and its activity within tumors by endoscopic ultrasound elastography. RESULTS: VCN-01 replicated in PDAC models and exerted antitumor effects which were improved when combined with chemotherapy. Hyaluronidase expression by VCN-01 degraded tumor stroma and facilitated delivery of a variety of therapeutic agents such as chemotherapy and therapeutic antibodies. Clinically, treatment was generally well-tolerated and resulted in disease stabilization of injected lesions. VCN-01 was detected in blood as secondary peaks and in post-treatment tumor biopsies, indicating virus replication. Patients had increasing levels of hyaluronidase in sera over time and decreased tumor stiffness, suggesting stromal disruption. CONCLUSIONS: VCN-01 is an oncolytic adenovirus with direct antitumor effects and stromal disruption capabilities, representing a new therapeutic agent for cancers with dense stroma. TRIAL REGISTRATION NUMBER: EudraCT number: 2012-005556-42 and NCT02045589.


Assuntos
Adenoviridae/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Células Estromais/efeitos dos fármacos , Albuminas/administração & dosagem , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Feminino , Humanos , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Gencitabina
11.
Int J Hyperthermia ; 26(5): 456-64, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20569110

RESUMO

PURPOSE: In controlled laboratory studies of hyperthermia and thermal ablation, translucent hydrogels containing bovine serum albumin (BSA) are often employed as tissue-mimicking materials due to the change in their opacity that takes place as they accumulate heat damage. In this work we demonstrate the biological relevance of this optical metric of thermal damage, as well as establish the physical mechanisms that link it with quantifiable damage to the proteins embedded in the gel. MATERIALS AND METHODS: We applied Fourier transform infrared (FTIR) spectroscopy, turbidity analysis using ultraviolet-visible (UV/VIS) spectroscopy, and size exclusion chromatography (SEC) to samples of heat-treated, aqueous bovine serum albumin (BSA). We also measured the rates of survival in heated suspensions of breast cancer cells using a colorimetric assay. RESULTS: Using FTIR spectroscopy and SEC, we show that the intermolecular beta-sheet content of the protein ensemble rises in heat treatments above 60 degrees C, which causes aggregate formation. Furthermore, by applying UV/VIS spectroscopy we demonstrate that the opacity of the hydrogel increases past 60 degrees C due to the formation of insoluble protein aggregates that scatter incident light. Finally, we illustrate that the viability of human breast cancer cells follows a similar trend to measurements of BSA polyacrylamide hydrogel opacity at various temperatures from 37 degrees C to 90 degrees C. CONCLUSIONS: Our work establishes a causal link between the degree of BSA denaturation in hydrogel and the opacity of the medium. Furthermore, our results demonstrate that BSA hydrogels provide a simple physical model for quantifying biologically relevant heat damage in real time during controlled laboratory studies of hyperthermia and thermal ablation.


Assuntos
Temperatura Alta , Hidrogéis/química , Soroalbumina Bovina/química , Resinas Acrílicas , Animais , Neoplasias da Mama , Bovinos , Linhagem Celular Tumoral , Sobrevivência Celular , Cromatografia em Gel , Feminino , Humanos , Nefelometria e Turbidimetria , Desnaturação Proteica , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier
12.
J Acoust Soc Am ; 128(5): EL310-15, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21110544

RESUMO

Poor drug penetration through tumor tissue has emerged as a fundamental obstacle to cancer therapy. The aim of this study was to examine the ability of cavitation instigated by high-intensity focused ultrasound (HIFU) to increase convective transport of a model therapeutic in an in vitro tumor model. Cavitation activity was quantified by analyzing passively recorded acoustic emissions, and mass transfer was quantified using post-treatment image analysis of the distribution of a dye-labeled macromolecule. The strong correlation between cavitation activity and drug delivery suggests the potential for non-invasive treatment and monitoring.


Assuntos
Antineoplásicos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Substâncias Macromoleculares/farmacocinética , Neoplasias/tratamento farmacológico , Terapia por Ultrassom/métodos , Acústica , Arteriopatias Oclusivas/diagnóstico por imagem , Humanos , Técnicas In Vitro , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Microambiente Tumoral , Ultrassonografia
13.
Sci Transl Med ; 11(476)2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30674657

RESUMO

Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.


Assuntos
Adenoviridae/fisiologia , Terapia de Alvo Molecular , Vírus Oncolíticos/fisiologia , Proteína do Retinoblastoma/metabolismo , Retinoblastoma/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Camundongos , Metástase Neoplásica , Coelhos , Retinoblastoma/imunologia , Retinoblastoma/patologia , Análise de Sobrevida , Distribuição Tecidual , Pesquisa Translacional Biomédica , Resultado do Tratamento , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Clin Cancer Res ; 21(6): 1406-18, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25391696

RESUMO

PURPOSE: Tumor targeting upon intravenous administration and subsequent intratumoral virus dissemination are key features to improve oncolytic adenovirus therapy. VCN-01 is a novel oncolytic adenovirus that combines selective replication conditional to pRB pathway deregulation, replacement of the heparan sulfate glycosaminoglycan putative-binding site KKTK of the fiber shaft with an integrin-binding motif RGDK for tumor targeting, and expression of hyaluronidase to degrade the extracellular matrix. In this study, we evaluate the safety and efficacy profile of this novel oncolytic adenovirus. EXPERIMENTAL DESIGN: VCN-01 replication and potency were assessed in a panel of tumor cell lines. VCN-01 tumor-selective replication was evaluated in human fibroblasts and pancreatic islets. Preclinical toxicity, biodistribution, and efficacy studies were conducted in mice and Syrian hamsters. RESULTS: Toxicity and biodistribution preclinical studies support the selectivity and safety of VCN-01. Antitumor activity after intravenous or intratumoral administration of the virus was observed in all tumor models tested, including melanoma and pancreatic adenocarcinoma, both in immunodeficient mice and immunocompetent hamsters. CONCLUSIONS: Oncolytic adenovirus VCN-01 characterized by the expression of hyaluronidase and the RGD shaft retargeting ligand shows an efficacy-toxicity prolife in mice and hamsters by intravenous and intratumoral administration that warrants clinical testing.


Assuntos
Adenocarcinoma/terapia , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animais , Sítios de Ligação/genética , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Matriz Extracelular/metabolismo , Feminino , Células HEK293 , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Hialuronoglucosaminidase/biossíntese , Hialuronoglucosaminidase/genética , Ilhotas Pancreáticas/virologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/genética , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Control Release ; 169(1-2): 40-7, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23562636

RESUMO

Oncolytic viruses (OV) and ultrasound-enhanced drug delivery are powerful novel technologies. OV selectively self-amplify and kill cancer cells but their clinical use has been restricted by limited delivery from the bloodstream into the tumor. Ultrasound has been previously exploited for targeted release of OV in vivo, but its use to induce cavitation, microbubble oscillations, for enhanced OV tumor extravasation and delivery has not been previously reported. By identifying and optimizing the underlying physical mechanism, this work demonstrates that focused ultrasound significantly enhances the delivery and biodistribution of systemically administered OV co-injected with microbubbles. Up to a fiftyfold increase in tumor transgene expression was achieved, without any observable tissue damage. Ultrasound exposure parameters were optimized as a function of tumor reperfusion time to sustain inertial cavitation, a type of microbubble activity, throughout the exposure. Passive detection of acoustic emissions during treatment confirmed inertial cavitation as the mechanism responsible for enhanced delivery and enabled real-time monitoring of successful viral delivery.


Assuntos
Adenoviridae/fisiologia , Sistemas de Liberação de Medicamentos/instrumentação , Neoplasias/terapia , Terapia Viral Oncolítica/instrumentação , Vírus Oncolíticos/fisiologia , Ultrassom/instrumentação , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Desenho de Equipamento , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microbolhas , Neoplasias/genética , Vírus Oncolíticos/genética , Transgenes
16.
J Natl Cancer Inst ; 105(22): 1701-10, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24168971

RESUMO

BACKGROUND: Oncolytic viruses are among the most powerful and selective cancer therapeutics under development and are showing robust activity in clinical trials, particularly when administered directly into tumor nodules. However, their intravenous administration to treat metastatic disease has been stymied by unfavorable pharmacokinetics and inefficient accumulation in and penetration through tumors. METHODS: Adenovirus (Ad) was "stealthed" with a new N-(2-hydroxypropyl)methacrylamide polymer, and circulation kinetics were characterized in Balb/C SCID mice (n = 8 per group) bearing human ZR-75-1 xenograft tumors. Then, to noninvasively increase extravasation of the circulating polymer-coated Ad into the tumor, it was coinjected with gas microbubbles and the tumor was exposed to 0.5 MHz focused ultrasound at peak rarefactional pressure of 1.2 MPa. These ultrasound exposure conditions were designed to trigger inertial cavitation, an acoustic phenomenon that produces shock waves and can be remotely monitored in real-time. Groups were compared with Student t test or one-way analysis of variance with Tukey correction where groups were greater than two. All statistical tests were two-sided. RESULTS: Polymer-coating of Ad reduced hepatic sequestration, infection (>8000-fold; P < .001), and toxicity and improved circulation half-life (>50-fold; P = .001). Combination of polymer-coated Ad, gas bubbles, and focused ultrasound enhanced tumor infection >30-fold; (4 × 10(6) photons/sec/cm(2); standard deviation = 3 × 10(6) with ultrasound vs 1.3 × 10(5); standard deviation = 1 × 10(5) without ultrasound; P = .03) and penetration, enabling kill of cells more than 100 microns from the nearest blood vessel. This led to substantial and statistically significant retardation of tumor growth and increased survival. CONCLUSIONS: Combining drug stealthing and ultrasound-induced cavitation may ultimately enhance the efficacy of a range of powerful therapeutics, thereby improving the treatment of metastatic cancer.


Assuntos
Acrilamidas/administração & dosagem , Adenoviridae , Adjuvantes Farmacêuticos/administração & dosagem , Neoplasias da Mama/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Veículos Farmacêuticos/administração & dosagem , Terapia por Ultrassom , Análise de Variância , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Terapia Combinada/métodos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Microbolhas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Control Release ; 157(2): 235-42, 2012 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-21982902

RESUMO

We investigated whether ultrasound-induced cavitation at 0.5 MHz could improve the extravasation and distribution of a potent breast cancer-selective oncolytic adenovirus, AdEHE2F-Luc, to tumour regions that are remote from blood vessels. We developed a novel tumour-mimicking model consisting of a gel matrix containing human breast cancer cells traversed by a fluid channel simulating a tumour blood vessel, through which the virus and microbubbles could be made to flow. Ultrasonic pressures were chosen to maximize either broadband emissions, associated with inertial cavitation, or ultraharmonic emissions, associated with stable cavitation, while varying duty cycle to keep the total acoustic energy delivered constant for comparison across exposures. None of the exposure conditions tested affected cell viability in the absence of the adenovirus. When AdEHE2F-Luc was delivered via the vessel, inertial cavitation increased transgene expression in tumour cells by up to 200 times. This increase was not observed in the absence of Coxsackie and Adenovirus Receptor cell expression, discounting sonoporation as the mechanism of action. In the presence of inertial cavitation, AdEHE2F-Luc distribution was greatly improved in the matrix surrounding the vessel, particularly in the direction of the ultrasound beam; this enabled AdEHE2F-Luc to kill up to 80% of cancer cells within the ultrasound focal volume in the gel 24 hours after delivery, compared to 0% in the absence of cavitation.


Assuntos
Acústica , Adenoviridae , Neoplasias da Mama/diagnóstico por imagem , Sistemas de Liberação de Medicamentos , Animais , Neoplasias da Mama/terapia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos , Ultrassonografia
18.
Cancer Res ; 72(19): 5060-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22915754

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is characterized by the combined occurrence of pituitary, pancreatic, and parathyroid tumors showing loss of heterozygosity in the putative tumor suppressor gene MEN1. This gene encodes the protein menin, the overexpression of which inhibits cell proliferation in vitro. In this study, we conducted a preclinical evaluation of MEN1 gene therapy in pituitary tumors of Men1(+/-) mice, using a recombinant nonreplicating adenoviral serotype 5 vector that contained the murine Men1 cDNA under control of a cytomegalovirus promoter (Men1.rAd5). Pituitary tumors in 55 Men1(+/-) female mice received a transauricular intratumoral injection of Men1.rAd5 or control treatments, followed by 5-bromo-2-deoxyuridine (BrdUrd) in drinking water for four weeks before magnetic resonance imaging (MRI) and immunohistochemical analysis. Immediate procedure-related and 4-week mortalities were similar in all groups, indicating that the adenoviral gene therapy was not associated with a higher mortality. Menin expression was higher in the Men1.rAd5-treated mice when compared with other groups. Daily proliferation rates assessed by BrdUrd incorporation were reduced significantly in Men1.rAd5-injected tumors relative to control-treated tumors. In contrast, apoptotic rates, immune T-cell response, and tumor volumes remained similar in all groups. Our findings establish that MEN1 gene replacement therapy can generate menin expression in pituitary tumors, and significantly reduce tumor cell proliferation.


Assuntos
Adenoma/terapia , Proliferação de Células , Modelos Animais de Doenças , Terapia Genética/métodos , Neoplasias Hipofisárias/terapia , Proteínas Proto-Oncogênicas/genética , Adenoma/genética , Adenoma/metabolismo , Adenoviridae/genética , Animais , Vetores Genéticos/genética , Células HEK293 , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipófise/diagnóstico por imagem , Hipófise/metabolismo , Hipófise/patologia , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Radiografia
19.
Ultrasound Med Biol ; 37(11): 1838-52, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21963037

RESUMO

A flow-through tissue-mimicking phantom composed of a biocompatible hydro-gel with embedded tumour cells was used to assess and optimize the role of ultrasound-induced cavitation on the extravasation of a macromolecular compound from a channel mimicking vessel in the gel, namely a non-replicating luciferase-expressing adenovirus (Ad-Luc). Using a 500 KHz therapeutic ultrasound transducer confocally aligned with a focussed passive cavitation detector, different exposure conditions and burst mode timings were selected by performing time and frequency domain analysis of passively recorded acoustic emissions, in the absence and in the presence of ultrasound contrast agents acting as cavitation nuclei. In the presence of Sonovue, maximum ultraharmonic emissions were detected for peak rarefactional pressures of 360 kPa, and maximum broadband emissions occurred at 1250 kPa. The energy of the recorded acoustic emissions was used to optimise the pulse repetition frequency and duty cycle in order to maximize either ultraharmonic or broadband emissions while keeping the acoustic energy delivered to the focus constant. Cell viability measurements indicated that none of the insonation conditions investigated induces cell death in the absence of a therapeutic agent (i.e. virus). Phase contrast images of the tissue-mimicking phantom showed that short range vessel disruption can occur when ultra-harmonic emissions (nf0/2) are maximised whereas formation of a micro-channel perpendicular to the flow can be obtained in the presence of broadband acoustic emissions. Following Ad-Luc delivery, luciferase expression measurements showed that a 60-fold increase in its bioavailability can be achieved when broadband noise emissions are present during insonation, even for modest contrast agent concentrations. The findings of the present study suggest that drug delivery systems based on acoustic cavitation may help enhance the extravasation of anticancer agents, thus increasing their penetration distance to hypoxic regions and poorly vascularised tumour regions.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Sonicação , Adenoviridae , Animais , Materiais Biocompatíveis , Sobrevivência Celular , Meios de Contraste/química , Desenho de Equipamento , Extravasamento de Materiais Terapêuticos e Diagnósticos , Ondas de Choque de Alta Energia , Hidrogéis , Técnicas In Vitro , Luciferases , Substâncias Macromoleculares/química , Camundongos , Microbolhas , Imagens de Fantasmas , Fosfolipídeos/química , Hexafluoreto de Enxofre/química , Transdutores , Células Tumorais Cultivadas
20.
J Neurosci Res ; 85(2): 332-41, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17086548

RESUMO

Several neurotrophic factors, including brain-derived neurotrophic factor (BDNF), and neurotransmitters, such as glutamate, may influence neuronal apoptotic death. Rat cerebellar granule neurons (CGN) cultured in low potassium (5 or 10 mM KCl) for more than 5 days in vitro (DIV) die apoptotically. These cells survive in the presence of high potassium (25 mM KCl, K25) or N-methyl-D-aspartate (NMDA), an agonist of glutamatergic receptors. CGN transferred from high to low potassium die apoptotically. Here, we characterized the effect of BDNF and NMDA on the apoptotic death induced by low potassium in CGN. Cell death of CGN by culturing in low potassium for 6 DIV was inhibited by BDNF and NMDA. When CGN were cultured in K25 and transferred to a low-potassium medium, 65% of neurons died after 48 hr. Under these conditions, BDNF, NMDA, or BDNF + NMDA increased CGN survival. Both BDNF and NMDA decreased caspase-9 activity and mRNA caspase-3 levels and activity induced by low potassium. CGN survival induced by BDNF is mediated by TrkB activation, whereas that induced by NMDA is mediated by NMDA receptor and TrkB activation. NMDA, but not BDNF, raised [Ca(2+)](i), which was reduced by low-potassium treatment. These results suggest that NMDA receptor stimulation induces CGN survival through the influx of extracellular Ca(2+) that may evoke the release of BDNF and the activation of TrkB. Complementary mechanisms induced by depolarization and changes in Ca(2+) levels would also contribute to the neuroprotection exerted by NMDA and potassium.


Assuntos
Apoptose/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Potássio/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Cerebelo/metabolismo , Cerebelo/patologia , Immunoblotting , Neurônios/fisiologia , RNA Mensageiro/análise , Ratos , Receptor trkB/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA