Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cancer Immunol Res ; 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38819256

RESUMO

Chimeric antigen receptor (CAR) T cells express an extracellular domain consisting of a single-chain fragment variable (scFv) targeting a surface tumor-associated antigen. scFv selection should involve safety profiling with evaluation of the efficacy/toxicity balance, especially when the target antigen also is expressed on healthy cells. Here, to assess differences in terms of efficacy and on-target/off-tumor effects, we five different CARs targeting CD123 by substituting only the scFv. In in vitro models, T cells engineered to express three of these five CD123 CARs were effectively cytotoxic on leukemic cells without increasing lysis of monocytes or endothelial cells. Using the IncuCyte® system, we confirmed the low cytotoxicity of CD123 CAR T cells on endothelial cells. Hematotoxicity evaluation using progenitor culture and CD34 cell lysis showed that two of the five CD123 CAR T cells were less cytotoxic on hematopoietic stem cells. Using a humanized mouse model, we confirmed that CD123- cells were not eliminated by the CD123 CAR T cells. Two CD123 CAR T cells reduced tumor infiltration and increased overall survival of mice in three in vivo models of blastic plasmacytoid dendritic cell neoplasm. In an aggressive version of this model, bulk RNA sequencing analysis showed that these CD123 CAR T cells upregulated genes associated with cytotoxicity and activation/exhaustion a few days after the injection. Together, these results emphasize the importance of screening different scFvs for the development of CAR constructs to support selection of cells with the optimal risk-benefit ratio for clinical development.

2.
J Immunother Cancer ; 10(7)2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35803613

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) remains a very difficult disease to cure due to the persistence of leukemic stem cells (LSCs), which are resistant to different lines of chemotherapy and are the basis of refractory/relapsed (R/R) disease in 80% of patients with AML not receiving allogeneic transplantation. METHODS: In this study, we showed that the interleukin-1 receptor accessory protein (IL-1RAP) protein is overexpressed on the cell surface of LSCs in all subtypes of AML and confirmed it as an interesting and promising target in AML compared with the most common potential AML targets, since it is not expressed by the normal hematopoietic stem cell. After establishing the proof of concept for the efficacy of chimeric antigen receptor (CAR) T-cells targeting IL-1RAP in chronic myeloid leukemia, we hypothesized that third-generation IL-1RAP CAR T-cells could eliminate AML LSCs, where the medical need is not covered. RESULTS: We first demonstrated that IL-1RAP CAR T-cells can be produced from AML T-cells at the time of diagnosis and at relapse. In vitro and in vivo, we showed the effectiveness of IL-1RAP CAR T-cells against AML cell lines expressing different levels of IL-1RAP and the cytotoxicity of autologous IL-1RAP CAR T-cells against primary cells from patients with AML at diagnosis or at relapse. In patient-derived relapsed AML xenograft models, we confirmed that IL-1RAP CAR T-cells are able to circulate in peripheral blood and to migrate in the bone marrow and spleen, are cytotoxic against primary AML cells and increased overall survival. CONCLUSION: In conclusion, our preclinical results suggest that IL-1RAP CAR T-based adoptive therapy could be a promising strategy in AML treatment and it warrants the clinical investigation of this CAR T-cell therapy.


Assuntos
Leucemia Mieloide Aguda , Receptores de Antígenos Quiméricos , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imunoterapia , Proteína Acessória do Receptor de Interleucina-1/metabolismo , Leucemia Mieloide Aguda/terapia , Recidiva , Linfócitos T
3.
Curr Protoc Mouse Biol ; 9(2): e62, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31145554

RESUMO

Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. While curative approaches for early stage HCC exist, effective treatment options for advanced HCC are lacking. Furthermore, there are no efficient chemopreventive strategies to limit HCC development once cirrhosis is established. One challenge for drug development is unsatisfactory animal models. In this article, we describe an orthotopic xenograft mouse model of human liver cancer cell lines through image-guided injection into the liver. This technique provides a less invasive yet highly efficient approach to engraft human HCC into mouse liver. Similarly, image-guided injections are used to deliver chemotherapeutics locally, enabling reduction in potential systemic adverse effects, while reducing the required dose for a therapeutic effect. In summary, this image-guided strategy provides a novel and convenient approach to improve current HCC mouse models. © 2019 The Authors. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.


Assuntos
Xenoenxertos/fisiologia , Neoplasias Hepáticas Experimentais/terapia , Camundongos , Transplante Heterólogo/métodos , Ultrassom/métodos , Animais , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Transplante Heterólogo/instrumentação , Ultrassom/instrumentação
4.
Gut ; 67(4): 736-745, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28360099

RESUMO

OBJECTIVE: HCV infection is a leading cause of chronic liver disease and a major indication for liver transplantation. Although direct-acting antivirals (DAAs) have much improved the treatment of chronic HCV infection, alternative strategies are needed for patients with treatment failure. As an essential HCV entry factor, the tight junction protein claudin-1 (CLDN1) is a promising antiviral target. However, genotype-dependent escape via CLDN6 and CLDN9 has been described in some cell lines as a possible limitation facing CLDN1-targeted therapies. Here, we evaluated the clinical potential of therapeutic strategies targeting CLDN1. DESIGN: We generated a humanised anti-CLDN1 monoclonal antibody (mAb) (H3L3) suitable for clinical development and characterised its anti-HCV activity using cell culture models, a large panel of primary human hepatocytes (PHH) from 12 different donors, and human liver chimeric mice. RESULTS: H3L3 pan-genotypically inhibited HCV pseudoparticle entry into PHH, irrespective of donor. Escape was likely precluded by low surface expression of CLDN6 and CLDN9 on PHH. Co-treatment of a panel of PHH with a CLDN6-specific mAb did not enhance the antiviral effect of H3L3, confirming that CLDN6 does not function as an entry factor in PHH from multiple donors. H3L3 also inhibited DAA-resistant strains of HCV and synergised with current DAAs. Finally, H3L3 cured persistent HCV infection in human-liver chimeric uPA-SCID mice in monotherapy. CONCLUSIONS: Overall, these findings underscore the clinical potential of CLDN1-targeted therapies and describe the functional characterisation of a humanised anti-CLDN1 antibody suitable for further clinical development to complement existing therapeutic strategies for HCV.


Assuntos
Anticorpos Monoclonais/farmacologia , Antivirais/farmacologia , Claudina-1/antagonistas & inibidores , Hepacivirus/efeitos dos fármacos , Hepatite C/prevenção & controle , Hepatócitos/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Animais , Claudina-1/imunologia , Hepatite C/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Camundongos , Camundongos SCID , Resultado do Tratamento
5.
Sci Rep ; 7(1): 13935, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29066853

RESUMO

Hepatocellular carcinoma (HCC) is the only cancer for which non-invasive diagnosis is recognized by international guidelines. Contrast agent free ultrasound imaging, computed tomography (CT) and/or magnetic resonance imaging are techniques used for early detection and confirmation. Clinical evidence depicts that CT is 30% less precise as compared to MRI for detection of small tumors. In our work, we have reported some novel tools that can enhance the sensitivity and precision of CT applied to preclinical research (micro-CT). Our system, containing non-toxic nano-droplets loaded with iodine has high contrasting properties, liver and hepatocyte specificity and strong liver persistence. Micro-CT was performed on HCC model implanted in nude mice by intrahepatic injection. Contrast agent was administrated intravenously. This method allows an unprecedented high precision of detection, quantitative measurement of tumor volume and quantitative follow-up of the tumor development.


Assuntos
Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/patologia , Halogenação , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Fígado/diagnóstico por imagem , Nanotecnologia , Microtomografia por Raio-X/métodos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Emulsões , Feminino , Humanos , Fígado/patologia , Camundongos , Ultrassonografia
6.
Glob Chall ; 1(4): 1700013, 2017 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-31565271

RESUMO

Medical application of siRNAs relies on methods for delivering nucleic acids into the cytosol. Synthetic carriers, which assemble with nucleic acids into delivery systems, show promises for cancer therapy but efficiency remains to be improved. In here, the effectiveness of pyridylthiourea-polyethylenimine (πPEI), a siRNA carrier that favors both polyplex disassembly and endosome rupture upon sensing the acidic endosomal environment, in 3 experimental models of hepatocellular cancer is tested. The πPEI-assisted delivery of a siRNA targeting the polo-like kinase 1 into Huh-7 monolayer produces a 90% cell death via a demonstrated RNA interference mechanism. Incubation of polyplex with Huh-7 spheroids leads to siRNA delivery into the superficial first cell layer and a 60% reduction in spheroid growth compared to untreated controls. Administration of polyplexes into mice bearing subcutaneous implanted Huh-7Luc tumors results in a reduced tumor progression, similar to the one observed in the spheroid model. Altogether, these results support the in vivo use of synthetic and dedicated polymers for increasing siRNA-mediated gene knockdown, and their clinical promise in cancer therapeutics.

7.
Sci Rep ; 6: 35230, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27739457

RESUMO

The development of multimodal strategies for the treatment of hepatocellular carcinoma requires tractable animal models allowing for advanced in vivo imaging. Here, we characterize an orthotopic hepatocellular carcinoma model based on the injection of luciferase-expressing human hepatoma Huh-7 (Huh-7-Luc) cells in immunodeficient mice. Luciferase allows for an easy repeated monitoring of tumor growth by in vivo bioluminescence. The intrahepatic injection was more efficient than intrasplenic or intraportal injection in terms of survival, rate of orthotopic engraftment, and easiness. A positive correlation between luciferase activity and tumor size, evaluated by Magnetic Resonance Imaging, allowed to define the endpoint value for animal experimentation with this model. Response to standard of care, sorafenib or doxorubicin, were similar to those previously reported in the literature, with however a strong toxicity of doxorubicin. Tumor vascularization was visible by histology seven days after Huh-7-Luc transplantation and robustly developed at day 14 and day 21. The model was used to explore different imaging modalities, including microtomography, probe-based confocal laser endomicroscopy, full-field optical coherence tomography, and ultrasound imaging. Tumor engraftment was similar after echo-guided intrahepatic injection as after laparotomy. Collectively, this orthotopic hepatocellular carcinoma model enables the in vivo evaluation of chemotherapeutic and surgical approaches using multimodal imaging.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Luciferases/metabolismo , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Imagem Multimodal/métodos , Transplante de Neoplasias/patologia , Ultrassonografia/métodos
8.
Nanoscale ; 8(13): 7240-7, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26974603

RESUMO

"Pop goes the particle". Here we report on the preparation of redox responsive mesoporous organo-silica nanoparticles containing disulfide (S-S) bridges (ss-NPs) that, even upon the exohedral grafting of targeting ligands, retained their ability to undergo structural degradation, and increase their local release activity when exposed to a reducing agent. This degradation could be observed also inside glioma C6 cancer cells. Moreover, when anticancer drug-loaded pristine and derivatized ss-NPs were fed to glioma C6 cells, the responsive hybrids were more effective in their cytotoxic action compared to non-breakable particles. The possibility of tailoring the surface functionalization of this hybrid, yet preserving its self-destructive behavior and enhanced drug delivery properties, paves the way for the development of effective biodegradable materials for in vivo targeted drug delivery.


Assuntos
Dissulfetos/química , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Dióxido de Silício/química , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/farmacocinética , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Humanos , Oxirredução/efeitos dos fármacos , Tamanho da Partícula , Porosidade , Substâncias Redutoras/farmacologia , Temozolomida
9.
Mol Med Rep ; 13(3): 2645-54, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26820174

RESUMO

Adoptive immunotherapy using autologous cytokine-induced killer (CIK) cells reduces the recurrence rate of hepatocellular carcinoma (HCC) in association with transarterial chemoembolization or radiofrequency. However, a large­scale development of this immunotherapy remains difficult to consider in an autologous setting, considering the logistical hurdles associated with the production of this cell therapy product. A previous study has provided the in vitro and in vivo proof­of­concept that allogeneic suicide gene­modified killer cells (aSGMKCs) from healthy blood donors (a cell therapy product previously demonstrated to provide anti­leukemic effects to patients receiving allogeneic hematopoietic transplantation) may exert a potent anti­tumor effect towards HCC. Therefore, the development of a bank of 'ready­for­use' aSGMKCs was proposed as an approach allowing for the development of immunotherapies that are more convenient and on a broader scale than that of autologous therapies. In the present study, aSGMKCs were compared with CIK cells generated according to three different protocols. Similar to CIK cells, the cytotoxic activity of aSGMKCs toward the Huh­7 HCC cell line was mediated by tumor necrosis factor­related apoptosis­inducing ligand, tumor necrosis factor­α and interferon­Î³. Furthermore, the frequency of natural killer (NK), NK­like T and T cells, and their in vitro and in vivo cytotoxicity activities were similar between aSGMKCs and CIK cells. Thus, the present study demonstrated that aSGMKCs are similar to CIK cells, further suggesting the possibility for future use of aSGMKCs in the treatment of solid tumors, including HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Células Matadoras Induzidas por Citocinas/imunologia , Genes Transgênicos Suicidas , Neoplasias Hepáticas/terapia , Animais , Linhagem Celular Tumoral , Células Matadoras Induzidas por Citocinas/transplante , Testes Imunológicos de Citotoxicidade , Feminino , Células HeLa , Humanos , Imunoterapia Adotiva , Masculino , Camundongos , Fenótipo , Transplante Homólogo
10.
Ann Surg Oncol ; 23(Suppl 5): 567-573, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26511264

RESUMO

BACKGROUND: Tumor-specific fluorescent antibodies, which can be recognized at a cellular or tissue level using optical imaging such as confocal laser endomicroscopy (CLE), could provide a means for rapid and accurate tumor diagnosis and staging. The aim of this study was to evaluate the ability of CLE to detect the presence of tagged cells within lymph nodes in an original simulated metastatic model. MATERIALS AND METHODS: A solution of indocyanine green containing a suspension of porcine hepatocytes, marked with carboxy-fluorescein-succinimidyl-ester (CFSE), was injected endoscopically in the gastric submucosa of 10 pigs. Fluorescence lymphography using a near-infrared laparoscope was used to identify sentinel and secondary drainage nodes. Additionally, a nonfluorescent gastric and a mesenteric node were identified. Every 5-10 min, those nodes were scanned using probe-based or needle-based CLE (pCLE or nCLE). Immunohistochemistry (IHC) using anti-cytokeratin 18 antibodies was subsequently performed to confirm the presence of hepatocytes in the lymph nodes. RESULTS: A total of 36 lymph nodes were analyzed with both CLE probes. Hepatocyte penetration in lymph nodes, as assessed by repeated CLE scanning, took 10-40 min after submucosal injection. Concordance between CLE and IHC was 84 and 72 % for pCLE and nCLE, respectively. False negatives were partly due to incomplete CFSE labeling of hepatocytes, which could not be recognized by CLE, but were detected with IHC. CONCLUSIONS: Real-time CLE analysis effectively recognized the presence in perigastric nodes of marked hepatic cells that had been injected endoscopically in the stomach. Validation studies on tumor-bearing animals using tumor-specific antibodies should be performed.


Assuntos
Imuno-Histoquímica/métodos , Linfonodo Sentinela/diagnóstico por imagem , Linfonodo Sentinela/patologia , Animais , Anticorpos , Corantes , Feminino , Mucosa Gástrica , Hepatócitos/imunologia , Verde de Indocianina , Queratina-18/imunologia , Laparoscopia/métodos , Metástase Linfática , Masculino , Mesentério , Microscopia Confocal/métodos , Modelos Biológicos , Estudo de Prova de Conceito , Suínos
11.
Hum Gene Ther Methods ; 26(6): 197-210, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26467420

RESUMO

We evaluated the possibility of introducing a combination of six oncogenes into primary porcine hepatocytes (PPH) using a lentiviral vector (LV)-mediated gene transfer in order to develop a porcine hepatocellular carcinoma model based on autologous transplantation of ex vivo-transformed hepatocytes. The six oncogenes were introduced into three plasmids, hence enabling the production of LVs encoding a luciferase reporter gene and hTERT+p53(DD), cyclinD1+CDK4(R24C), and c-myc(T58A)+HRas(G21V) genes, respectively. In order to improve the protection of the laboratory personnel manipulating such LVs, we used a compact cell culture cassette (CliniCell(®) device) as a closed cell culture system. We demonstrated that the CliniCell device allows to produce LVs, through plasmid transfection of 293T cells, and, after transfer to a second cassette, to transduce PPH with a similar efficacy as conventional open cell culture systems such as flasks or Petri dishes. Additionally, it is possible to cryopreserve at -80°C the transduced cells, directly in the CliniCell device used for the transduction. In conclusion, the use of a closed culture system for the safe handling of oncogene-encoding LVs lays the foundation for the development of porcine tumor models based on the autologous transplantation of ex vivo-transformed primary cells.


Assuntos
Técnicas de Cultura Celular por Lotes , Vetores Genéticos/genética , Vetores Genéticos/isolamento & purificação , Lentivirus/genética , Replicação Viral , Animais , Técnicas de Cultura Celular por Lotes/instrumentação , Técnicas de Cultura Celular por Lotes/métodos , Técnicas de Cultura Celular por Lotes/normas , Expressão Gênica , Genes Reporter , Células HEK293 , Hepatócitos , Humanos , Suínos , Transdução Genética , Transgenes
13.
Biochem Biophys Res Commun ; 465(4): 658-64, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26241675

RESUMO

High-risk human papillomavirus (HPV) types 16 and 18 are associated with more than 70% of cervical cancer cases. The oncoprotein E6 is multifunctional and has numerous cellular partners. The best-known activity of E6 is the polyubiquination of the pro-apoptotic tumor suppressor p53, targeting it for degradation by the 26S proteasome. Loss of p53 triggers genomic instability and favors cancer development. Here, we generated recombinant adenovirus (Ad) vectors expressing artificial microRNAs directed against HPV16 E6 (Ad16_1) or HPV18 E6 (Ad18_2). E6-knockdown was observed in HeLa after treatment with Ad18_2 and in SiHa with Ad16_1. Western-blot experiments found an increase in p53 levels after treatment in both cell lines. Cell death was observed in both cell lines after knockdown of E6. Further analysis such as cleavage of caspases (3 and 7) as well as of PARP1 indicated that treated HeLa and SiHa cells underwent apoptosis. The growth of HeLa-derived tumors developed in nude mice was significantly reduced after intra-tumoral injection of Ad18_2. Therefore, vectorisation of artificial miRNA against E6 oncoprotein by means of recombinant adenoviruses might represent a valuable therapeutic approach for treating HPV-positive cancers.


Assuntos
Apoptose/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , MicroRNAs/genética , Proteínas Oncogênicas Virais/antagonistas & inibidores , Proteínas Oncogênicas Virais/genética , Infecções por Papillomavirus/terapia , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Neoplasias do Colo do Útero/terapia , Adenoviridae/genética , Animais , Linhagem Celular , Feminino , Técnicas de Silenciamento de Genes , Terapia Genética , Vetores Genéticos , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidade , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/patogenicidade , Humanos , Camundongos , Camundongos Nus , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nat Biotechnol ; 33(5): 549-554, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25798937

RESUMO

Hepatitis C virus (HCV) infection is a leading cause of liver cirrhosis and cancer. Cell entry of HCV and other pathogens is mediated by tight junction (TJ) proteins, but successful therapeutic targeting of TJ proteins has not been reported yet. Using a human liver-chimeric mouse model, we show that a monoclonal antibody specific for the TJ protein claudin-1 (ref. 7) eliminates chronic HCV infection without detectable toxicity. This antibody inhibits HCV entry, cell-cell transmission and virus-induced signaling events. Antibody treatment reduces the number of HCV-infected hepatocytes in vivo, highlighting the need for de novo infection by means of host entry factors to maintain chronic infection. In summary, we demonstrate that an antibody targeting a virus receptor can cure chronic viral infection and uncover TJ proteins as targets for antiviral therapy.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Claudina-1/imunologia , Hepatite C/terapia , Cirrose Hepática/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Claudina-1/uso terapêutico , Hepacivirus/imunologia , Hepacivirus/patogenicidade , Hepatite C/imunologia , Hepatite C/virologia , Hepatócitos/imunologia , Humanos , Cirrose Hepática/terapia , Cirrose Hepática/virologia , Camundongos
15.
Surg Innov ; 22(3): 217-22, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25801192

RESUMO

INTRODUCTION: Image fusion between ultrasound (US) and computed tomography (CT) scan or magnetic resonance can increase operator accuracy in targeting liver lesions, particularly when those are undetectable with US alone. We have developed a modular gel to simulate hepatic solid lesions for educational purposes in imaging and minimally invasive ablation techniques. We aimed to assess the impact of image fusion in targeting artificial hepatic lesions during the hands-on part of 2 courses (basic and advanced) in hepatobiliary surgery. MATERIALS AND METHODS: Under US guidance, 10 fake tumors of various sizes were created in the livers of 2 pigs, by percutaneous injection of a biocompatible gel engineered to be hyperdense on CT scanning and barely detectable on US. A CT scan was obtained and a CT-US image fusion was performed using the ACUSON S3000 US system (Siemens Healthcare, Germany). A total of 12 blinded course attendants, were asked in turn to perform a 10-minute liver scan with US alone followed by a 10-minute scan using image fusion. RESULTS: Using US alone, the expert managed to identify all lesions successfully. The true positive rate for course attendants with US alone was 14/36 and 2/24 in the advanced and basic courses, respectively. The total number of false positives identified was 26. With image fusion, the rate of true positives significantly increased to 31/36 (P < .001) in the advanced group and 16/24 in the basic group (P < .001). The total number of false positives, considering all participants, decreased to 4 (P < .001). CONCLUSIONS: Image fusion significantly increases accuracy in targeting hepatic lesions and might improve echo-guided procedures.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Ultrassonografia de Intervenção/métodos , Animais , Fígado/diagnóstico por imagem , Neoplasias Hepáticas/diagnóstico por imagem , Modelos Biológicos , Imagens de Fantasmas , Suínos
16.
Gut ; 64(3): 483-94, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24848265

RESUMO

OBJECTIVE: Although direct-acting antiviral agents (DAAs) have markedly improved the outcome of treatment in chronic HCV infection, there continues to be an unmet medical need for improved therapies in difficult-to-treat patients as well as liver graft infection. Viral entry is a promising target for antiviral therapy. DESIGN: Aiming to explore the role of entry inhibitors for future clinical development, we investigated the antiviral efficacy and toxicity of entry inhibitors in combination with DAAs or other host-targeting agents (HTAs). Screening a large series of combinations of entry inhibitors with DAAs or other HTAs, we uncovered novel combinations of antivirals for prevention and treatment of HCV infection. RESULTS: Combinations of DAAs or HTAs and entry inhibitors including CD81-, scavenger receptor class B type I (SR-BI)- or claudin-1 (CLDN1)-specific antibodies or small-molecule inhibitors erlotinib and dasatinib were characterised by a marked and synergistic inhibition of HCV infection over a broad range of concentrations with undetectable toxicity in experimental designs for prevention and treatment both in cell culture models and in human liver-chimeric uPA/SCID mice. CONCLUSIONS: Our results provide a rationale for the development of antiviral strategies combining entry inhibitors with DAAs or HTAs by taking advantage of synergy. The uncovered combinations provide perspectives for efficient strategies to prevent liver graft infection and novel interferon-free regimens.


Assuntos
Antivirais/uso terapêutico , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/administração & dosagem , Linhagem Celular , Quimera , Sinergismo Farmacológico , Quimioterapia Combinada , Hepatite C/prevenção & controle , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Camundongos , Camundongos SCID
17.
Mol Ther ; 22(3): 634-644, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24445938

RESUMO

Cell therapy based on alloreactivity has completed clinical proof of concept against hematological malignancies. However, the efficacy of alloreactivity as a therapeutic approach to treat solid tumors is unknown. Using cell culture and animal models, we aimed to investigate the efficacy and safety of allogeneic suicide gene-modified killer cells as a cell-based therapy for hepatocellular carcinoma (HCC), for which treatment options are limited. Allogeneic killer cells from healthy donors were isolated, expanded, and phenotypically characterized. Antitumor cytotoxic activity and safety were studied using a panel of human or murine HCC cell lines engrafted in immunodeficient or immunocompetent mouse models. Human allogeneic suicide gene-modified killer cells (aSGMKCs) exhibit a high, rapid, interleukin-2-dependent, and non-major histocompatibility complex class I-restricted in vitro cytotoxicity toward human hepatoma cells, mainly mediated by natural killer (NK) and NK-like T cells. In vivo evaluation of this cell therapy product demonstrates a marked, rapid, and sustained regression of HCC. Preferential liver homing of effector cells contributed to its marked efficacy. Calcineurin inhibitors allowed preventing rejection of allogeneic lymphocytes by the host immune system without impairing their antitumor activity. Our results demonstrate proof of concept for aSGMKCs as immunotherapy for HCC and open perspectives for the clinical development of this approach.


Assuntos
Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/terapia , Citotoxicidade Imunológica , Neoplasias Hepáticas/imunologia , Transplante de Neoplasias/imunologia , Linfócitos T/imunologia , Transplante Homólogo/métodos , Animais , Linhagem Celular Tumoral , Células HeLa , Humanos , Imunoterapia Adotiva , Neoplasias Hepáticas/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Linfócitos T/transplante
18.
Front Microbiol ; 4: 213, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23898329

RESUMO

Hepatitis C virus (HCV) is a major cause of cirrhosis and hepatocellular carcinoma (HCC) making the virus the most common cause of liver failure and transplantation. HCV is estimated to chronically affect 130 million individuals and to lead to more than 350,000 deaths per year worldwide. A vaccine is currently not available. The recently developed direct acting antivirals (DAAs) have markedly increased the efficacy of the standard of care but are not efficient enough to completely cure all chronically infected patients and their toxicity limits their use in patients with advanced liver disease, co-morbidity or transplant recipients. Because of the host restriction, which is limited to humans and non-human primates, in vivo study of HCV infection has been hampered since its discovery more than 20 years ago. The chimpanzee remains the most physiological model to study the innate and adaptive immune responses, but its use is ethically difficult and is now very restricted and regulated. The development of a small animal model that allows robust HCV infection has been achieved using chimeric liver immunodeficient mice, which are therefore not suitable for studying the adaptive immune responses. Nevertheless, these models allowed to go deeply in the comprehension of virus-host interactions and to assess different therapeutic approaches. The immunocompetent mouse models that were recently established by genetic humanization have shown an interesting improvement concerning the study of the immune responses but are still limited by the absence of the complete robust life cycle of the virus. In this review, we will focus on the relevant available animal models of HCV infection and their usefulness for deciphering the HCV life cycle and virus-induced liver disease, as well as for the development and evaluation of new therapeutics. We will also discuss the perspectives on future immunocompetent mouse models and the hurdles to their development.

19.
J Mater Chem B ; 1(36): 4692-4700, 2013 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32261213

RESUMO

The present study describes hybrid nanoparticles, built by alternate deposition of siRNA and modified polyethyleneimine (tyrosine-grafted PEI or tyrosine/galactose-grafted PEI) on calcium phosphate nanoparticles. These "easy to produce" nanoparticles (NPs) present an efficient gene silencing effect demonstrated in vitro in a luciferase expressing cell culture model and in vivo in a tumour xenograft mouse model. The luciferase gene silencing percentage reached up to 95% in vitro with biocompatible doses of siRNA. Interestingly, we show by SPECT imaging of radiolabeled particles that without modifying the size, stability and in vitro efficiency, the grafting of a sugar moiety on PEI can modify the in vivo biodistribution of the particles. The proof of concept that galactose-grafting on PEI could change biodistribution without changing the gene silencing efficiency makes them versatile tools for specific delivery of small interfering RNA. As they have been designed so far, biodistribution is mainly located in the liver and thus these innovative nanoparticles open a realistic and feasible strategy for siRNA delivery into the liver in vivo.

20.
J Hepatol ; 55(3): 718-720, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21616105

RESUMO

BACKGROUND & AIMS: Studies of hepatitis C virus (HCV) infection, immunopathogenesis, and resulting liver diseases have been hampered by the lack of a small animal model. We developed humanized mice with human immune system and liver tissues to improve the studies of hepatitis C pathogenesis and treatment. METHODS: To promote engraftment of human hepatocytes, we expressed a fusion protein of the FK506 binding protein (FKBP) and caspase 8 under the control of the albumin promoter (AFC8), which induces liver cell death, in Balb/C Rag2(-/-) γC-null mice. Co-transplantation of human CD34(+) human hematopoietic stem cells (HSC) and hepatocyte progenitors into the transgenic mice led to efficient engraftment of human leucocytes and hepatocytes. We then infected these humanized mice (AFC8-hu HSC/Hep) with primary HCV isolates and studied HCV-induced immune responses and liver diseases. RESULTS: AFC8-hu HSC/Hep mice supported HCV infection in the liver and generated a human immune T-cell response against HCV. HCV infection induced liver inflammation, hepatitis, and fibrosis, which correlated with activation of stellate cells and expression of human fibrogenic genes. CONCLUSIONS: AFC8-hu HSC/Hep mice are a useful model of HCV infection, the immune response, and liver disease, because they contain human immune system and liver cells. These mice become infected with HCV, generate a specific immune response against the virus, and develop liver diseases that include hepatitis and fibrosis. This model might also be used to develop therapeutics for HCV infection.


Assuntos
Modelos Animais de Doenças , Transplante de Células-Tronco Hematopoéticas , Hepacivirus , Hepatite C/imunologia , Camundongos , Animais , Hepatite C/patologia , Hepatócitos/transplante , Humanos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA