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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-35163605

RESUMO

Due to their ease of isolation and their properties, mesenchymal stem cells (MSCs) have been widely investigated. MSCs have been proved capable of migration towards areas of inflammation, including tumors. Therefore, they have been suggested as vectors to carry therapies, specifically to neoplasias. As most of the individuals joining clinical trials that use MSCs for cancer and other pathologies are carefully recruited and do not suffer from other diseases, here we decided to study the safety and application of iv-injected MSCs in animals simultaneously induced with different inflammatory pathologies (diabetes, wound healing and tumors). We studied this by in vitro and in vivo approaches using different gene reporters (GFP, hNIS, and f-Luc) and non-invasive techniques (PET, BLI, or fluorescence). Our results found that MSCs reached different organs depending on the previously induced pathology. Moreover, we evaluated the property of MSCs to target tumors as vectors to deliver adenoviruses, including the interaction between tumor microenvironment and MSCs on their arrival. Mechanisms such as transdifferentiation, MSC fusion with cells, or paracrine processes after MSCs homing were studied, increasing the knowledge and safety of this new therapy for cancer.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neoplasias , Microambiente Tumoral , Animais , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/terapia
2.
J Immunol ; 191(9): 4589-98, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24062490

RESUMO

The ErbB network is dysregulated in many solid tumors. To exploit this, we have developed a chimeric Ag receptor (CAR) named T1E28z that targets several pathogenetically relevant ErbB dimers. T1E28z is coexpressed with a chimeric cytokine receptor named 4αß (combination termed T4), enabling the selective expansion of engineered T cells using IL-4. Human T4(+) T cells exhibit antitumor activity against several ErbB(+) cancer types. However, ErbB receptors are also expressed in several healthy tissues, raising concerns about toxic potential. In this study, we have evaluated safety of T4 immunotherapy in vivo using a SCID beige mouse model. We show that the human T1E28z CAR efficiently recognizes mouse ErbB(+) cells, rendering this species suitable to evaluate preclinical toxicity. Administration of T4(+) T cells using the i.v. or intratumoral routes achieves partial tumor regression without clinical or histopathologic toxicity. In contrast, when delivered i.p., tumor reduction is accompanied by dose-dependent side effects. Toxicity mediated by T4(+) T cells results from target recognition in both tumor and healthy tissues, leading to release of both human (IL-2/IFN-γ) and murine (IL-6) cytokines. In extreme cases, outcome is lethal. Both toxicity and IL-6 release can be ameliorated by prior macrophage depletion, consistent with clinical data that implicate IL-6 in this pathogenic event. These data demonstrate that CAR-induced cytokine release syndrome can be modeled in mice that express target Ag in an appropriate distribution. Furthermore, our findings argue that ErbB-retargeted T cells can achieve therapeutic benefit in the absence of unacceptable toxicity, providing that route of administration and dose are carefully optimized.


Assuntos
Imunoterapia Adotiva , Neoplasias/imunologia , Proteínas Oncogênicas v-erbB/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T/metabolismo , Animais , Linhagem Celular , Humanos , Interferon gama/biossíntese , Interferon gama/metabolismo , Interleucina-2/biossíntese , Interleucina-2/metabolismo , Interleucina-4 , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Macrófagos , Camundongos , Camundongos SCID , Neoplasias/terapia , Transdução de Sinais
3.
BMC Med ; 11: 139, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23710709

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) have been promoted as an attractive option to use as cellular delivery vehicles to carry anti-tumor agents, owing to their ability to home into tumor sites and secrete cytokines. Multiple isolated populations have been described as MSCs, but despite extensive in vitro characterization, little is known about their in vivo behavior.The aim of this study was to investigate the efficacy and efficiency of different MSC lineages derived from five different sources (bone marrow, adipose tissue, epithelial endometrium, stroma endometrium, and amniotic membrane), in order to assess their adequacy for cell-based anti-tumor therapies. Our study shows the crucial importance of understanding the interaction between MSCs and tumor cells, and provides both information and a methodological approach, which could be used to develop safer and more accurate targeted therapeutic applications. METHODS: We first measured the in vivo migration capacity and effect on tumor growth of the different MSCs using two imaging techniques: (i) single-photon emission computed tomography combined with computed tomography (SPECT-CT), using the human sodium iodine symporter gene (hNIS) and (ii) magnetic resonance imaging using superparamagnetic iron oxide. We then sought correlations between these parameters and expression of pluripotency-related or migration-related genes. RESULTS: Our results show that migration of human bone marrow-derived MSCs was significantly reduced and slower than that obtained with the other MSCs assayed and also with human induced pluripotent stem cells (hiPSCs). The qPCR data clearly show that MSCs and hiPSCs exert a very different pluripotency pattern, which correlates with the differences observed in their engraftment capacity and with their effects on tumor growth. CONCLUSION: This study reveals differences in MSC recruitment/migration toward the tumor site and the corresponding effects on tumor growth. Three observations stand out: 1) tracking of the stem cell is essential to check the safety and efficacy of cell therapies; 2) the MSC lineage to be used in the cell therapy needs to be carefully chosen to balance efficacy and safety for a particular tumor type; and 3) different pluripotency and mobility patterns can be linked to the engraftment capacity of the MSCs, and should be checked as part of the clinical characterization of the lineage.


Assuntos
Movimento Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/diagnóstico por imagem , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/diagnóstico por imagem , Neoplasias/diagnóstico por imagem , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/cirurgia , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
4.
J Clin Immunol ; 31(4): 710-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21505816

RESUMO

Adoptive immunotherapy using chimeric antigen receptor-engrafted T cells is a promising emerging therapy for cancer. Prior to clinical testing, it is mandatory to evaluate human therapeutic cell products in meaningful in vivo pre-clinical models. Here, we describe the use of fused single-photon emission CT-CT imaging to monitor real-time migration of chimeric antigen receptor-engineered T cells in immune compromised (SCID Beige) mice. Following intravenous administration, human T cells migrate in a highly similar manner to that reported in man, but penetrate poorly into established tumors. By contrast, when delivered via intraperitoneal or subcutaneous routes, T cells remain at the site of inoculation with minimal systemic absorption-irrespective of the presence or absence of tumor. Together, these data support the validity of pre-clinical testing of human T-cell immunotherapy in SCID Beige mice. In light of their established efficacy, regional administration of engineered human T cells represents an attractive therapeutic option to minimize toxicity in the treatment of selected malignancies.


Assuntos
Movimento Celular , Imunoterapia Adotiva/efeitos adversos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/imunologia , Transferência Adotiva/métodos , Animais , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/terapia , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos SCID , Mucina-1/imunologia , Proteínas Recombinantes de Fusão , Linfócitos T/citologia , Linfócitos T/transplante , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
EJNMMI Phys ; 8(1): 68, 2021 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-34626239

RESUMO

BACKGROUND: This study aimed to evaluate the performance of a preclinical PET insert in three configurations: as a stand-alone unit outside the MRI bore, inside the bore of a cryogen-free 3T MRI and, finally, while performing simultaneous PET/MRI studies. METHODS: The PET insert consists of two rings of six detectors, each detector comprising 8 × 12 SiPMs reading out dual offset layers of pixelated LYSO crystals with a 1.4-mm pitch. The inner diameter is 60 mm, transaxial field of view (FoV) 40 mm and axial FoV 98 mm. Evaluation was based on NEMA NU 4-2008 guidelines with appropriate modifications. Spatial resolution and sensitivity were measured inside and outside the MR bore. Image quality, count rate and quantitative performance were measured in all three configurations. The effect of temperature stability on PET sensitivity during fast spin echo sequences was also evaluated. B0 field homogeneity and T1 and T2 relaxation times were measured using a water-filled phantom, with and without simultaneous PET operation. Finally, PET and MRI scans of a mouse injected with 10 MBq [18F]NaF and a mouse injected with 16 MBq [18F]FDG were performed in sequential and simultaneous modes. RESULTS: Peak absolute sensitivity was 10.15% with an energy window of 250-750 keV. Absolute sensitivity values outside and inside the MR bore with MR idle agreed to within 0.1%. Outside the MR bore, spatial resolution was 1.21/1.59 mm FWHM (radial/tangential) 5 mm from the centre of the FoV which compared well with 1.19/1.26 mm FWHM inside the MR bore. There were no substantial differences between all three scan configurations in terms of peak NEC rate (175 kcps at 17 MBq), scatter or random fractions. Uniformity and recovery coefficients were also consistent between scanning modes. B0 field homogeneity and T1 and T2 relaxation times were unaltered by the presence of the PET insert. No significant differences were observed between sequential and simultaneous scans of the animals. CONCLUSIONS: We conclude that the performance of the PET insert and MRI system is not significantly affected by the scanning mode.

6.
Appl Radiat Isot ; 96: 91-101, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25479439

RESUMO

We evaluated and compared a new bombesin analog [Tyr-Gly5, Nle(14)]-BBN(6-14) conjugated to DOTA or DTPA and radiolabeled with In-111 in low and high GRPR expressing tumor models. Both peptides were radiolabeled with high radiochemical purity and specific activity. In vitro assays on T-47D, LNCaP and PC-3 cells showed that the affinity of peptides is similar and a higher binding and internalization of DOTA-peptide to PC-3 cells was observed. Both peptides could target PC-3 and LNCaP tumors in vivo and both tumor types could be visualized by microSPECT/CT.


Assuntos
Bombesina/análogos & derivados , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/metabolismo , Compostos Heterocíclicos com 1 Anel , Ácido Pentético/análogos & derivados , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo , Compostos Radiofarmacêuticos , Receptores da Bombesina/metabolismo , Animais , Bombesina/química , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Feminino , Xenoenxertos , Humanos , Técnicas In Vitro , Radioisótopos de Índio , Masculino , Camundongos SCID , Transplante de Neoplasias , Cintilografia , Compostos Radiofarmacêuticos/química , Distribuição Tecidual
7.
PLoS One ; 8(2): e57263, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451195

RESUMO

Interferon alpha (IFNα) is used for the treatment of hepatitis C infection and whilst efficacious it is associated with multiple adverse events including reduced leukocyte, erythrocyte, and platelet counts, fatigue, and depression. These events are most likely caused by systemic exposure to interferon. We therefore hypothesise that targeting the therapeutic directly to the intended site of action in the liver would reduce exposure in blood and peripheral tissue and hence improve the safety and tolerability of IFNα therapy. We genetically fused IFN to a domain antibody (dAb) specific to a hepatocyte restricted antigen, asialoglycoprotein receptor (ASGPR). Our results show that the murine IFNα2 homolog (mIFNα2) fused to an ASGPR specific dAb, termed DOM26h-196-61, could be expressed in mammalian tissue culture systems and retains the desirable biophysical properties and activity of both fusion partners when measured in vitro. Furthermore a clear increase in in vivo targeting of the liver by mIFNα2-ASGPR dAb fusion protein, compared to that observed with either unfused mIFNα2 or mIFNα2 fused to an isotype control dAb VHD2 (which does not bind ASGPR) was demonstrated using microSPECT imaging. We suggest that these findings may be applicable in the development of a liver-targeted human IFN molecule with improved safety and patient compliance in comparison to the current standard of care, which could ultimately be used as a treatment for human hepatitis virus infections.


Assuntos
Autoanticorpos/imunologia , Interferon-alfa/farmacologia , Fígado/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Autoanticorpos/química , Humanos , Interferon-alfa/imunologia , Camundongos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
8.
PLoS One ; 8(9): e73260, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023846

RESUMO

The αvß6 integrin is up-regulated in cancer and wound healing but it is not generally expressed in healthy adult tissue. There is increasing evidence that it has a role in cancer progression and will be a useful target for antibody-directed cancer therapies. We report a novel recombinant diabody antibody fragment that targets specifically αvß6 and blocks its function. The diabody was engineered with a C-terminal hexahistidine tag (His tag), expressed in Pichia pastoris and purified by IMAC. Surface plasmon resonance (SPR) analysis of the purified diabody showed affinity in the nanomolar range. Pre-treatment of αvß6-expressing cells with the diabody resulted in a reduction of cell migration and adhesion to LAP, demonstrating biological function-blocking activity. After radio-labeling, using the His-tag for site-specific attachment of (99m)Tc, the diabody retained affinity and targeted specifically to αvß6-expressing tumors in mice bearing isogenic αvß6 +/- xenografts. Furthermore, the diabody was specifically internalized into αvß6-expressing cells, indicating warhead targeting potential. Our results indicate that the new αvß6 diabody has a range of potential applications in imaging, function blocking or targeted delivery/internalization of therapeutic agents.


Assuntos
Antígenos de Neoplasias/imunologia , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/imunologia , Integrinas/imunologia , Engenharia de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Animais , Especificidade de Anticorpos , Antígenos de Neoplasias/metabolismo , Linhagem Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fragmentos de Imunoglobulinas/química , Integrinas/metabolismo , Marcação por Isótopo , Camundongos , Pichia/genética , Proteínas Recombinantes/química , Neoplasias Cutâneas/metabolismo , Tecnécio/química
9.
Clin Cancer Res ; 15(21): 6595-601, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19861465

RESUMO

PURPOSE: The Na/I symporter (hNIS) promotes concentration of iodine in cells. In cancer gene therapy, this transgene has potential as a reporter gene for molecular imaging of viral biodistribution and as a therapeutic protein promoting (131)I-mediated radiotherapy. Here, we combined the imaging and therapeutic potential of hNIS in an oncolytic adenoviruses targeting colorectal cancer cells. EXPERIMENTAL DESIGN: We generated an adenovirus (AdIP2) encoding hNIS and capable of selective replication in colorectal carcinoma cells. The selectivity of this virus was verified in vitro and in vivo. Its spread in tumors was monitored in vivo using single-photon emission computed tomography/CT imaging upon (99m)TcO(4)(-) injection and confirmed by immunohistochemistry. Metabolic radiotherapy was done through injection of therapeutic doses of (131)I(-). RESULTS: We showed in vitro and in vivo the selectivity of AdIP2 and that hNIS expression is restricted to the target cells. Imaging and immunohistochemical data showed that viral spread is limited and that the point of maximal hNIS expression is reached 48 hours after a single intratumoral injection. Administration of a single therapeutic dose of (131)I at this time point led to a dramatic reduction in tumor size not observed in hNIS-negative viruses. CONCLUSIONS: This report showed for the first time that the combination of the imaging and therapeutic potentials of hNIS can be applied to oncolytic adenoviruses in experimental models of cancer.


Assuntos
Adenoviridae/genética , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/terapia , Genes Reporter , Terapia Genética/métodos , Radioisótopos do Iodo/uso terapêutico , Simportadores/genética , Proteínas Wnt/metabolismo , Animais , Linhagem Celular Tumoral , Diagnóstico por Imagem , Marcação de Genes , Vetores Genéticos , Camundongos , Camundongos Nus , Cintilografia , Simportadores/metabolismo , Transgenes
10.
Cancer Res ; 69(6): 2655-62, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19258509

RESUMO

The lack of safe and efficient systemic gene delivery vectors has largely reduced the potential of gene therapy in the clinic. Previously, we have reported that polypropylenimine dendrimer PPIG3/DNA nanoparticles are capable of tumor transfection upon systemic administration in tumor-bearing mice. To be safely applicable in the clinic, it is crucial to investigate the colloidal stability of nanoparticles and to monitor the exact biodistribution of gene transfer in the whole body of the live subject. Our biophysical characterization shows that dendrimers, when complexed with DNA, are capable of forming spontaneously in solution a supramolecular assembly that possesses all the features required to diffuse in experimental tumors through the enhanced permeability and retention effect. We show that these nanoparticles are of sizes ranging from 33 to 286 nm depending on the DNA concentration, with a colloidal stable and well-organized fingerprint-like structure in which DNA molecules are condensed with an even periodicity of 2.8 nm. Whole-body nuclear imaging using small-animal nano-single-photon emission computed tomography/computer tomography scanner and the human Na/I symporter (NIS) as reporter gene shows unique and highly specific tumor targeting with no detection of gene transfer in any of the other tissues of tumor-bearing mice. Tumor-selective transgene expression was confirmed by quantitative reverse transcription-PCR at autopsy of scanned animals, whereas genomic PCR showed that the tumor sites are the predominant sites of nanoparticle accumulation. Considering that NIS imaging of transgene expression has been recently validated in humans, our data highlight the potential of these nanoparticles as a new formulation for cancer gene therapy.


Assuntos
DNA/química , Técnicas de Transferência de Genes , Nanopartículas/química , Polipropilenos/química , Animais , Coloides/química , DNA/genética , Dendrímeros/química , Estabilidade de Medicamentos , Feminino , Análise de Fourier , Células HeLa , Humanos , Luz , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Nus , Microscopia Eletrônica de Transmissão/métodos , Plasmídeos/química , Plasmídeos/genética , Espalhamento de Radiação , Transplante Heterólogo
11.
Nat Med ; 14(2): 199-204, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18246079

RESUMO

Rhinoviruses cause serious morbidity and mortality as the major etiological agents of asthma exacerbations and the common cold. A major obstacle to understanding disease pathogenesis and to the development of effective therapies has been the lack of a small-animal model for rhinovirus infection. Of the 100 known rhinovirus serotypes, 90% (the major group) use human intercellular adhesion molecule-1 (ICAM-1) as their cellular receptor and do not bind mouse ICAM-1; the remaining 10% (the minor group) use a member of the low-density lipoprotein receptor family and can bind the mouse counterpart. Here we describe three novel mouse models of rhinovirus infection: minor-group rhinovirus infection of BALB/c mice, major-group rhinovirus infection of transgenic BALB/c mice expressing a mouse-human ICAM-1 chimera and rhinovirus-induced exacerbation of allergic airway inflammation. These models have features similar to those observed in rhinovirus infection in humans, including augmentation of allergic airway inflammation, and will be useful in the development of future therapies for colds and asthma exacerbations.


Assuntos
Modelos Animais de Doenças , Hipersensibilidade/virologia , Infecções por Picornaviridae/virologia , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Rhinovirus/fisiologia , Animais , Formação de Anticorpos/efeitos da radiação , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/virologia , Quimiocinas/biossíntese , Quimiocinas/imunologia , Fatores Quimiotáticos/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Humanos , Hipersensibilidade/imunologia , Imunidade Inata/efeitos da radiação , Inflamação , Mediadores da Inflamação/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Camundongos Transgênicos , Muco/metabolismo , Neutrófilos/imunologia , Neutrófilos/efeitos da radiação , Sistema Respiratório/imunologia , Sistema Respiratório/efeitos da radiação , Rhinovirus/efeitos da radiação , Células Th1/imunologia , Células Th1/efeitos da radiação , Células Th2/imunologia , Células Th2/efeitos da radiação , Raios Ultravioleta , Inativação de Vírus/efeitos da radiação , Replicação Viral/efeitos da radiação
12.
J Gen Virol ; 84(Pt 10): 2829-2836, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-13679617

RESUMO

Human rhinoviruses (HRV) are responsible for the majority of virus infections of the upper respiratory tract. Furthermore, HRV infection is associated with acute exacerbation of asthma and other chronic respiratory diseases of the lower respiratory tract. A small animal model of HRV-induced disease is required for the development of new therapies. However, existing mouse models of HRV infection are difficult to work with and until recently mouse cell lines were thought to be generally non-permissive for HRV replication in vitro. In this report we demonstrate that a virus of the minor receptor group, HRV1B, can infect and replicate in a mouse respiratory epithelial cell line (LA-4) more efficiently than in a mouse fibroblast cell line (L). The major receptor group virus HRV16 requires human intercellular adhesion molecule-1 (ICAM-1) for cell entry and therefore cannot infect LA-4 cells. However, transfection of in vitro-transcribed HRV16 RNA resulted in the replication of viral RNA and production of infectious virus. Expression of a chimeric ICAM-1 molecule, comprising mouse ICAM-1 with extracellular domains 1 and 2 replaced by the equivalent human domains, rendered the otherwise non-permissive mouse respiratory epithelial cell line susceptible to entry and efficient replication of HRV16. These observations suggest that the development of mouse models of respiratory tract infection by major as well as minor group HRV should be pursued.


Assuntos
Células Epiteliais/virologia , Sistema Respiratório/virologia , Rhinovirus/fisiologia , Replicação Viral , Animais , Linhagem Celular , Efeito Citopatogênico Viral , Células HeLa , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , RNA Viral/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sistema Respiratório/citologia , Rhinovirus/genética , Rhinovirus/patogenicidade , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA