Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Horm Metab Res ; 51(12): 805-811, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31826275

RESUMO

The whole world has been affected by a dramatically increasing prevalence of diabetes. Today, the etiology of both type 1 and type 2 diabetes is thought to revolve around the dysfunction of ß-cells, the insulin producing cells of the body. Within the pharmaceutical industry, the evaluation of new drugs for diabetes treatment is mostly done using cell lines or rodent islets and depends solely on the assessment of static insulin secretion. However, the use of cell lines or rodent islets is limiting lack of similarity of the human islet cells, leading to a constrain of the predictive value regarding the clinical potential of newly developed drugs. To overcome this issue, we developed an Engineered Micro-Pancreas as a unique platform for drug discovery. The Engineered Micro Pancreas is composed of (i) an organ-derived micro-scaffold, specifically a decellularized porcine lung-derived micro-scaffold and (ii) cadaveric islets seeded thereon. The Engineered Micro Pancreas remained viable and maintained insulin secretion in vitro for up to three months. The quantities of insulin were comparable to those secreted by freshly isolated human islets and therefore hold the potential for real-time and metabolic physiology mimicking drug screening.


Assuntos
Células Secretoras de Insulina/metabolismo , Pulmão/química , Pâncreas/metabolismo , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Sobrevivência Celular , Descoberta de Drogas , Matriz Extracelular/química , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Espectrometria de Massas , Pâncreas/crescimento & desenvolvimento , Suínos , Engenharia Tecidual/instrumentação
2.
Stem Cells Int ; 2017: 4061975, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28698719

RESUMO

Incurable neuroretinal degeneration diseases cause severe vision loss and blindness in millions of patients worldwide. In previous studies, we demonstrated that transplanting human bone marrow stromal cells (hBMSCs) in the extravascular spaces of the choroid (EVSC) of the Royal College of Surgeon rats ameliorated retinal degeneration for up to 5 months. Assessing the safety of hBMSC treatment and graft survival in a large animal is a crucial step before initiating clinical trials. Here, we transplanted hBMSCs into the EVSC compartment of New Zealand White rabbits. No immunosuppressants were used. Transplanted cells were spread across the EVSC covering over 80 percent of the subretinal surface. No cells were detected in the sclera. Cells were retained in the EVSC compartment 10 weeks following transplantation. Spectral domain optical coherence tomography (SD-OCT) and histopathology analysis demonstrated no choroidal hemorrhages, retinal detachment, inflammation, or any untoward pathological reactions in any of transplanted eyes or in the control noninjected contralateral eyes. No reduction in retinal function was recorded by electroretinogram up to 10 weeks following transplantation. This study demonstrates the feasibility and safety of transplanting hBMSCs in the EVSC compartment in a large eye model of rabbits.

3.
Stem Cell Res ; 15(2): 387-94, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26322852

RESUMO

Vision incapacitation and blindness associated with incurable retinal degeneration affect millions of people worldwide. In this study, 0.25×10(6) human bone marrow stem cells (hBM-MSCs) were transplanted epiretinally in the right eye of Royal College Surgeons (RCS) rats at the age of 28 days. Epiretinally transplanted cells were identified as a thin layer of cells along vitreous cavity, in close proximity to the retina or attached to the lens capsule, up to 6 weeks following transplantation. Epiretinal transplantation delayed photoreceptor degeneration and rescued retinal function up to 20 weeks following cell transplantation. Visual functions remained close to normal levels in epiretinal transplantation rats. No inflammation or any other adverse effects were observed in transplanted eyes. Our findings suggest that transplantation of hBM-MSCs as a thin epiretinal layer is effective for treatment of retinal degeneration in RCS rats, and that transplanting the cells in close proximity to the retina enhances hBM-MSC therapeutic effect compared with intravitreal injection.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Retina/fisiologia , Degeneração Retiniana/terapia , Adulto , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Modelos Animais de Doenças , Membrana Epirretiniana/metabolismo , Membrana Epirretiniana/patologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Microscopia de Fluorescência , Pessoa de Meia-Idade , Ratos , Transplante Heterólogo
4.
J Nanobiotechnology ; 13: 34, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25947109

RESUMO

BACKGROUND: Iron oxide (IO) nanoparticles (NPs) of sizes less than 50 nm are considered to be non-toxic, biodegradable and superparamagnetic. We have previously described the generation of IO NPs coated with Human Serum Albumin (HSA). HSA coating onto the IO NPs enables conjugation of the IO/HSA NPs to various biomolecules including proteins. Here we describe the preparation and characterization of narrow size distribution core-shell NIR fluorescent IO/HSA magnetic NPs conjugated covalently to Fibroblast Growth Factor 2 (FGF2) for biomedical applications. We examined the biological activity of the conjugated FGF2 on human bone marrow mesenchymal stem cells (hBM-MSCs). These multipotent cells can differentiate into bone, cartilage, hepatic, endothelial and neuronal cells and are being studied in clinical trials for treatment of various diseases. FGF2 enhances the proliferation of hBM-MSCs and promotes their differentiation toward neuronal, adipogenic and osteogenic lineages in vitro. RESULTS: The NPs were characterized by transmission electron microscopy, dynamic light scattering, ultraviolet-visible spectroscopy and fluorescence spectroscopy. Covalent conjugation of the FGF2 to the IO/HSA NPs significantly stabilized this growth factor against various enzymes and inhibitors existing in serum and in tissue cultures. IO/HSA NPs conjugated to FGF2 were internalized into hBM-MSCs via endocytosis as confirmed by flow cytometry analysis and Prussian Blue staining. Conjugated FGF2 enhanced the proliferation and clonal expansion capacity of hBM-MSCs, as well as their adipogenic and osteogenic differentiation to a higher extent compared with the free growth factor. Free and conjugated FGF2 promoted the expression of neuronal marker Microtubule-Associated Protein 2 (MAP2) to a similar extent, but conjugated FGF2 was more effective than free FGF2 in promoting the expression of astrocyte marker Glial Fibrillary Acidic Protein (GFAP) in these cells. CONCLUSIONS: These results indicate that stabilization of FGF2 by conjugating the IO/HSA NPs can enhance the biological efficacy of FGF2 and its ability to promote hBM-MSC cell proliferation and trilineage differentiation. This new system may benefit future therapeutic use of hBM-MSCs.


Assuntos
Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Nanopartículas de Magnetita , Células-Tronco Mesenquimais/citologia , Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Estabilidade de Medicamentos , Compostos Férricos/química , Fator 2 de Crescimento de Fibroblastos/química , Fluorescência , Humanos , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Tamanho da Partícula , Fotodegradação , Albumina Sérica/química , Espectrometria de Fluorescência
5.
Harefuah ; 154(2): 84-8, 138, 2015 Feb.
Artigo em Hebraico | MEDLINE | ID: mdl-25856858

RESUMO

BACKGROUND: Incurable retinal degenerations affect millions worldwide. Stem cell transplantation rescued visual functions in animal models of retinal degeneration. In those studies, cells were transplanted in subretinal "blebs". A limited number of cells could be injected and photoreceptor rescue was restricted to areas in proximity to the injection sites. PURPOSE: To develop a minimally-invasive surgical system for stem cell transplantation in the subretina and extravascular spaces of the choroid. METHODS: A novel syringe with flexible needle and adjustable pin was developed. Human bone marrow mesenchymal stem cells [hBM-MSCs) were transplanted in the eyes of RCS rats and NZW rabbits through a longitudinal triangular scleral incision. No immunosuppressants were used. Retinal function was determined by electroretinogram analysis and retinal structure was determined by histological analysis and optical coherence tomography (OCT). RESULTS: Transplanted cells were identified as a thin layer across the subretina and extravascular spaces of the choroid. In RCS rats, cell transplantation delayed photoreceptor degeneration across the entire retina and significantly enhanced retinal functions. No changes in retinal functions were recorded in rabbits following transplantation. No retinal detachment or choroidal hemorrhages were observed. CONCLUSIONS: The novel syringe facilitates cell transplantation across the subretina and extravascular spaces of the choroid using a minimally-invasive procedure. Human BM-MSC transplantation using this system ameliorates retinal degeneration in the animal model. DISCUSSION: This new transplantation system may increase the therapeutic effect of other cell-based therapies and therapeutic agents. This study is expected to lead directly to phase I clinical trials for autologous hBM-MSCs transplantation in patients with retinal degeneration.


Assuntos
Transplante de Medula Óssea/métodos , Corioide/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Degeneração Retiniana/cirurgia , Animais , Eletrorretinografia , Feminino , Humanos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Coelhos , Ratos , Tomografia de Coerência Óptica
6.
Exp Eye Res ; 118: 135-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24239509

RESUMO

Vision incapacitation and blindness associated with retinal degeneration affect millions of people worldwide. Cell based therapy and specifically transplantation of human adult bone marrow-derived stem cells (hBM-MSCs) present possible treatment strategy. Subretinal transplantation of human or rat BM-MSCs was shown previously to improve retinal function in Royal College Surgeons (RCS) rats. In those studies cells were transplanted via a transscleral-transchoroidal approach, creating a localized subretinal bleb. Limited number of cells could be injected and photoreceptor rescue was restricted to areas in proximity to the injection site. Here we describe a new surgical method for subretinal transplantation that facilitates uniform distribution of transplanted cells as a thin layer along most of the subretinal space. We assessed the therapeutic effect of hBM-MSCs on RCS rats when transplanted either subretinally or intravitreally. We also examined whether a second transplantation can prolong the therapeutic effect. A cell suspension of 2.5 × 10(6) cells in 5 µl was injected subretinally or intravitreally in RCS rats at 28 days postnatal. In the subretinal group, hBM-MSCs were transplanted posterior to the limbus in the superotemporal part of the eye through a longitudinal triangular scleral tunnel reaching the choroid. In the intravitreal group, the cells were injected into the superotemporal part of the vitreous cavity. In cross sections of subretinally transplanted eyes, removed 2 h following transplantation, hBM-MSCs were distributed as a near-homogenous thin layer along most of the subretinal space. In some animals the cells were also detected in the choroid. In the intravitreal injection group, hBM-MSCs were clustered in the vitreous cavity. Transplanted cells could be detected up to 2 weeks after transplantation but not at later time points. Retinal function and structure were assessed by electroretinogram (ERG) and histology analysis, respectively. Six weeks post transplantation, the mean maximal scotopic ERG b-wave amplitude response recorded in RCS control eyes was 1.2 µV. By contrast, in transplanted eyes mean responses of 56.4 µV and 66.2 µV were recorded in the intravitreally and subretinally transplanted eyes, respectively. In the subretinal group, retinal function was significantly higher in transplanted compared with control eyes up to 20 weeks following transplantation. By contrast, in the intravitreal group, rescue of retinal function persisted only up to 12 weeks following transplantation. Histological analysis revealed that 8 weeks following subretinal transplantation, the retinas of control eyes were dystrophic, with outer nuclear layer (ONL) containing a single cell layer. An extensive photoreceptor rescue was demonstrated in transplanted eyes at this time point, with 3-4 cell layers in the ONL along the entire retina. A second subretinal transplantation at 70 days postnatal did not enhance or prolong the therapeutic effect of hBM-MSCs. No immunosuppressants were used and long-term safety analysis demonstrated no gross or microscopic adverse effects. Taken together our findings suggest that transplantation of hBM-MSCs as a thin subretinal layer enhances the therapeutic effect and the safety of cell transplantation.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Degeneração Retiniana/cirurgia , Adulto , Animais , Modelos Animais de Doenças , Eletrorretinografia , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Ratos , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Resultado do Tratamento
7.
Isr Med Assoc J ; 16(12): 774-82, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25630208

RESUMO

BACKGROUND: Non-mobilized peripheral blood contains mostly committed cells with limited numbers of early progenitors. OBJECTIVES: To enrich functional progenitor cells from healthy donors and ischemic heart disease patients by short-term culture of mononuclear cells with defined culture conditions. METHODS: Mononuclear cells obtained from healthy donors and ischemic heart disease patients were cultured for7 days in a cytokine cocktail. We tested the multilineage differentiation capacities and phenotype of cultured cells. RESULTS: The short-term culture (7 days) of all study groups with a defined cytokine cocktail resulted in two distinct cell populations (adherent and non-adherent) that differed in their differentiation capacities as well as their cell surface markers. Cultured adherent cells showed higher differentiation potential and expressed endothelial and mesenchymal fibroblast-like surface markers as compared to fresh non-cultured mononuclear cells. The non-adherent cell fraction demonstrated high numbers of colony-forming units, indicating a higher differentiation potential of hematopoietic lineage. CONCLUSIONS: This study proved the feasibility of increasing limited numbers of multipotent progenitor cells obtained from the non-mobilized peripheral blood of healthy donors and ischemic patients. Moreover, we found that each of the two enriched subpopulations (adherent and non-adherent) has a different differentiation potential (mesenchymal, endothelial and hematopoietic).


Assuntos
Diferenciação Celular , Células Progenitoras Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Leucócitos Mononucleares , Células-Tronco Mesenquimais/fisiologia , Isquemia Miocárdica/sangue , Adulto , Técnicas de Cultura de Células , Células Cultivadas/metabolismo , Meios de Cultivo Condicionados , Citocinas/metabolismo , Estudos de Viabilidade , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Proteínas de Membrana/análise , Pessoa de Meia-Idade , Fatores de Tempo
8.
Clin Cancer Res ; 19(17): 4792-800, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23690483

RESUMO

PURPOSE: Adoptive cell transfer (ACT) using autologous tumor-infiltrating lymphocytes (TIL) was reported to yield objective responses in about 50% of metastatic patients with melanoma. Here, we present the intent-to-treat analysis of TIL ACT and analyze parameters predictive to response as well as the impact of other immunotherapies. EXPERIMENTAL DESIGN: Eighty patients with stage IV melanoma were enrolled, of which 57 were treated with unselected/young TIL and high-dose interleukin-2 (IL-2) following nonmyeloablative lymphodepleting conditioning. RESULTS: TIL cultures were established from 72 of 80 enrolled patients. Altogether 23 patients were withdrawn from the study mainly due to clinical deterioration during TIL preparation. The overall response rate and median survival was 29% and 9.8 months for enrolled patients and 40% and 15.2 months for treated patients. Five patients achieved complete and 18 partial remission. All complete responders are on unmaintained remission after a median follow-up of 28 months and the 3-year survival of responding patients was 78%. Multivariate analysis revealed blood lactate-dehydrogenase levels, gender, days of TIL in culture, and the total number of infused CD8+ cells as independent predictive markers for clinical outcome. Thirty-two patients received the CTLA-4-blocking antibody ipilimumab prior or post TIL infusion. Retrospective analysis revealed that nonresponders to ipilimumab or IL-2 based therapy had the same overall response rate to ACT as other patients receiving TIL. No additional toxicities to TIL therapy occurred following ipilimumab treatment. CONCLUSION: Adoptive transfer of TIL can yield durable and complete responses in patients with refractory melanoma, even when other immunotherapies have failed.


Assuntos
Transferência Adotiva/métodos , Terapia Baseada em Transplante de Células e Tecidos , Linfócitos do Interstício Tumoral , Melanoma/terapia , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Citotoxicidade Imunológica , Feminino , Humanos , Imunoterapia , Ipilimumab , Estimativa de Kaplan-Meier , Masculino , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/patologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias
9.
PLoS One ; 8(2): e57160, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451174

RESUMO

Vasculogenic mimicry (VM) describes functional vascular channels composed only of tumor cells and its presence predicts poor prognosis in melanoma patients. Inhibition of this alternative vascularization pathway might be of clinical importance, especially as several anti-angiogenic therapies targeting endothelial cells are largely ineffective in melanoma. We show the presence of VM structures histologically in a series of human melanoma lesions and demonstrate that cell cultures derived from these lesions form tubes in 3D cultures ex vivo. We tested the ability of nicotinamide, the amide form of vitamin B3 (niacin), which acts as an epigenetic gene regulator through unique cellular pathways, to modify VM. Nicotinamide effectively inhibited the formation of VM structures and destroyed already formed ones, in a dose-dependent manner. Remarkably, VM formation capacity remained suppressed even one month after the complete withdrawal of Nicotimamid. The inhibitory effect of nicotinamide on VM formation could be at least partially explained by a nicotinamide-driven downregulation of vascular endothelial cadherin (VE-Cadherin), which is known to have a central role in VM. Further major changes in the expression profile of hundreds of genes, most of them clustered in biologically-relevant clusters, were observed. In addition, nicotinamide significantly inhibited melanoma cell proliferation, but had an opposite effect on their invasion capacity. Cell cycle analysis indicated moderate changes in apoptotic indices. Therefore, nicotinamide could be further used to unravel new biological mechanisms that drive VM and tumor progression. Targeting VM, especially in combination with anti-angiogenic strategies, is expected to be synergistic and might yield substantial anti neoplastic effects in a variety of malignancies.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Melanoma/irrigação sanguínea , Neovascularização Patológica , Niacinamida/farmacologia , Vasos Sanguíneos/crescimento & desenvolvimento , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/patologia , Invasividade Neoplásica
10.
Immunotherapy ; 5(1): 79-90, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23256800

RESUMO

Immunotherapy holds a highly promising treatment approach for metastatic melanoma patients. Adoptive cell transfer (ACT) involves the ex vivo expansion of autologous antitumor reactive lymphocytes and their reinfusion into lymphodepleted patients, accompanied by IL-2 administration. ACT with tumor-infiltrating T lymphocytes demonstrates objective clinical responses in 50-72% of the patients, including 10-40% complete responses and was shown to produce durable disease control with long progression-free survival. Tumor-infiltrating T-lymphocyte ACT might even have curative potential as the vast majority of the complete responders are without any evidence of disease many years after treatment. Other adoptive transfer studies employ the genetic modification of T lymphocytes with genes encoding tumor-specific T cell receptors or antibody-based chimeric antigen receptors. These approaches opened numerous possibilities to treat cancers other than melanoma. In this article we will summarize the ACT strategies in melanoma, the new developments in this field and combinations with other therapies.


Assuntos
Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Humanos , Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/transplante , Melanoma/imunologia , Melanoma/mortalidade , Melanoma/terapia , Receptores de Antígenos de Linfócitos T/genética , Resultado do Tratamento
11.
J Immunother ; 34(2): 212-20, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21304398

RESUMO

Treatment of metastatic melanoma patients with adoptively transferred tumor infiltrating lymphocytes (TIL) has developed into an effective therapy. Various studies reported objective responses of 50% and more. The use of unselected, minimally cultured, bulk TIL (Young-TIL) has simplified the TIL production process and may therefore, allow the accessibility of this approach to cancer centers worldwide. This article describes the precise process leading to the large-scale production of Young-TIL for therapy. We have enrolled 55 melanoma patients and optimized their Young-TIL generation process. Young-TIL cultures were successfully established for 51 of 55 (93%) patients in 16.7 ± 5.5 days. In a large-scale expansion procedure Young-TIL of 32 patients were further expanded to treatment levels, resulting in a final number of 4.5 x 10¹° ± 2.0 x 10¹° TIL. Fifteen of 31 (48%) patients, who were evaluated, achieved a clinical response, including 4 complete and 11 partial responses. We confirmed the significant correlation between short culture duration, high number of infused cells, and tumor regression. A high percentage of CD8 T cells in the infusion product was beneficial to achieve an objective response. All responding patients were treated with Young-TIL cultures established in < 20 days. In summary, we describe here an efficient and reliable method to generate Young-TIL for adoptive transfer therapy, which may easily be adopted by other cancer centers and can lead to objective responses in 50% of refractory melanoma patients. In the future this approach may be used also in other types of malignancies.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia Adotiva , Linfócitos do Interstício Tumoral/citologia , Melanoma/terapia , Algoritmos , Células Cultivadas , Técnicas de Cocultura , Citotoxicidade Imunológica , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-2/farmacologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Melanoma/patologia , Melanoma/secundário , Estadiamento de Neoplasias , Resultado do Tratamento
12.
Clin Cancer Res ; 16(9): 2646-55, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20406835

RESUMO

PURPOSE: Adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TIL) has shown promising results in metastatic melanoma patients. Although objective response rates of over 50% have been reported, disadvantages of this approach are the labor-intensive TIL production and a very high drop-out rate of enrolled patients, limiting its widespread applicability. Previous studies showed a clear correlation between short TIL culture periods and clinical response. Therefore, we used a new TIL production technique using unselected, minimally cultured, bulk TIL (Young-TIL). The use of Young-TIL is not restricted to human leukocyte antigen (HLA)-A2 patients. The purpose of this study is to explore the efficacy and toxicity of adoptively transferred Young-TIL following lympho-depleting chemotherapy in metastatic melanoma patients, refractory to interleukin-2 and chemotherapy. EXPERIMENTAL DESIGN: Young-TIL cultures for 90% of the patients were successfully generated, enabling the treatment of most enrolled patients. We report here the results of 20 evaluated patients. RESULTS: Fifty percent of the patients achieved an objective clinical response according to the Response Evaluation Criteria in Solid Tumors, including two ongoing complete remissions (20+, 4+ months) and eight partial responses (progression-free survival: 18+, 13+, 10+, 9, 6+, 4, 3+, and 3 months). All responders are currently alive. Four additional patients showed disease stabilization. Side effects were transient and manageable. CONCLUSION: We showed that lympho-depleting chemotherapy followed by transfer of short-term cultured TIL can mediate tumor regression in 50% of metastatic melanoma with manageable toxicity. The convincing clinical results combined with the simplification of the process may thus have a major effect on cell therapy of cancer.


Assuntos
Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/transplante , Melanoma/terapia , Adulto , Idoso , Autoimunidade/imunologia , Células Cultivadas , Terapia Combinada , Diarreia/induzido quimicamente , Feminino , Seguimentos , Humanos , Imunoterapia Adotiva/efeitos adversos , Interleucina-2/efeitos adversos , Interleucina-2/uso terapêutico , Tempo de Internação , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Masculino , Melanoma/imunologia , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento
13.
Clin Dev Immunol ; 2010: 260267, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21234353

RESUMO

Adoptive Cell Transfer (ACT) of Tumor-Infiltrating Lymphocytes (TIL) in combination with lymphodepletion has proven to be an effective treatment for metastatic melanoma patients, with an objective response rate in 50%-70% of the patients. It is based on the ex vivo expansion and activation of tumor-specific T lymphocytes extracted from the tumor and their administration back to the patient. Various TIL-ACT trials, which differ in their TIL generation procedures and patient preconditioning, have been reported. In the latest clinical studies, genetically engineered peripheral T cells were utilized instead of TIL. Further improvement of adoptive T cell transfer depends on new investigations which seek higher TIL quality, increased durable response rates, and aim to treat more patients. Simplifying this therapy may encourage cancer centers worldwide to adopt this promising technology. This paper focuses on the latest progress regarding adoptive T cell transfer, comparing the currently available protocols and discussing their advantages, disadvantages, and implication in the future.


Assuntos
Imunoterapia Adotiva/métodos , Linfócitos do Interstício Tumoral/transplante , Melanoma/terapia , Linfócitos T/transplante , Ensaios Clínicos como Assunto , Humanos , Melanoma/imunologia , Resultado do Tratamento
14.
Cancer Immunol Immunother ; 59(2): 215-30, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19633846

RESUMO

It was previously shown that CEACAM1 on melanoma cells strongly predicts poor outcome. Here, we show a statistically significant increase of serum CEACAM1 in 64 active melanoma patients, as compared to 48 patients with no evidence of disease and 37 healthy donors. Among active patients, higher serum CEACAM1 correlated with LDH values and with decreased survival. Multivariate analysis with neutralization of LDH showed that increased serum CEACAM1 carries a hazard ratio of 2.40. In vitro, soluble CEACAM1 was derived from CEACAM1(+), but neither from CEACAM1(-) melanoma cells nor from CEACAM1(+) lymphocytes, and directly correlated with the number of CEACAM1(+) melanoma cells. Production of soluble CEACAM1 depended on intact de novo protein synthesis and secretion machineries, but not on metalloproteinase function. An unusually high percentage of CEACAM1(+) circulating NK and T lymphocytes was demonstrated in melanoma patients. CEACAM1 inhibited killing activity in functional assays. CEACAM1 expression could not be induced on lymphocytes by serum from patients with high CEACAM1 expression. Further, expression of other NK receptors was impaired, which collectively indicate on a general abnormality. In conclusion, the systemic dysregulation of CEACAM1 in melanoma patients further denotes the role of CEACAM1 in melanoma and may provide a basis for new tumor monitoring and prognostic platforms.


Assuntos
Antígenos CD/sangue , Antígenos CD/imunologia , Moléculas de Adesão Celular/sangue , Moléculas de Adesão Celular/imunologia , Melanoma/sangue , Melanoma/imunologia , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/imunologia , Adulto , Idoso , Feminino , Humanos , Células Matadoras Naturais/imunologia , Masculino , Pessoa de Meia-Idade , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Receptor 3 Desencadeador da Citotoxicidade Natural/imunologia , Receptores de IgG/imunologia , Linfócitos T/imunologia
15.
PLoS One ; 4(5): e5597, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19440333

RESUMO

BACKGROUND: NK cells are key players in anti tumor immune response, which can be employed in cell-based therapeutic modalities. One of the suggested ways to amplify their anti tumor effect, especially in the field of stem cell transplantation, is by selecting donor/recipient mismatches in specific HLA, to reduce the inhibitory effect of killer Ig-like receptors (KIRs). Here we suggest an alternative approach for augmentation of anti tumor effect of allogeneic NK cells, which is founded on profile matching of donor NK lysis receptors (NKLR) phenotype with tumor lysis-ligands. METHODOLOGY/PRINCIPAL FINDINGS: We show that an NKLR-mediated killing directly correlates with the NKLR expression intensity on NK cells. Considerable donor variability in the expression of CD16, NKp46, NKG2D and NKp30 on circulating NK cells, combined with the stability of phenotype in several independently performed tests over two months, indicates that NKLR-guided selection of donors is feasible. As a proof of concept, we show that melanoma cells are dominantly recognized by three NKLRs: NKG2D, NKp30 and NKp44. Notably, the expression of NKp30 on circulating NK cells among metastatic melanoma patients was significantly decreased, which diminishes their ability to kill melanoma cells. Ex vivo expansion of NK cells results not only in increased amount of cells but also in a consistently superior and predictable expression of NKG2D, NKp30 and NKp44. Moreover, expanded NK cultures with high expression of NKG2D or NKp30 were mostly derived from the corresponding NKG2D(high) or NK30(high) donors. These NK cultures subsequently displayed an improved cytotoxic activity against melanoma in a HLA/KIR-ligand mismatched setup, which was NKLR-dependent, as demonstrated with blocking anti-NKG2D antibodies. CONCLUSIONS/SIGNIFICANCE: NKLR/NKLR-ligand matching reproducibly elicits enhanced NK anti-tumor response. Common NKLR recognition patterns of tumors, as demonstrated here in melanoma, would allow implementation of this approach in solid malignancies and potentially in hematological malignancies, either independently or in adjunction to other modalities.


Assuntos
Células Matadoras Naturais/imunologia , Melanoma/imunologia , Receptores KIR/imunologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/metabolismo , Cinética , Melanoma/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores Imunológicos/imunologia , Células Tumorais Cultivadas
16.
J Immunother ; 32(4): 415-23, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19342963

RESUMO

Adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TIL) and high-dose interleukin-2 (IL-2), after nonmyeloablative chemotherapy, has been shown to result in tumor regression in half of refractory metastatic melanoma patients. In the present study, we describe 2 separate clinical protocols. Twelve patients were treated with "Selected"-TIL, as previously reported and 8 patients with the modified version of "Young"-TIL. Selected-TIL protocol required the establishment of multiple T-cell cultures from 1 patient and in vitro selection of cultures secreting interferon-gamma upon antigenic stimulation. In contrast, Young-TIL are minimally cultured T cells with superior in vitro features that do not require further selection. Two of 12 Selected-TIL patients experienced objective clinical responses (1 complete response, 1 partial response). Out of 8 treated Young-TIL patients, 1 experienced complete response, 2 partial response, and 4 patients had disease stabilization. Twenty-one of 33 enrolled Selected-TIL patients were excluded from the protocol, mainly as cultures failed the interferon-gamma selection criteria or due to clinical deterioration, compared with only 3 Young-TIL patients. Expected bone marrow suppression and high-dose IL-2 toxicity were transient. There was no treatment-related mortality. This study vindicates the feasibility and effectiveness of TIL technology and calls for further efforts to implement and enhance this modality. The use of minimally cultured, unselected Young-TIL enables the treatment of most enrolled patients. Although the cohort of Young-TIL patients treated so far is rather small and the follow-up short, the response rate is encouraging.


Assuntos
Imunoterapia Adotiva , Interleucina-2/uso terapêutico , Linfócitos do Interstício Tumoral/imunologia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Aciclovir/uso terapêutico , Adulto , Idoso , Antifúngicos/uso terapêutico , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/uso terapêutico , Antivirais/uso terapêutico , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Citotoxicidade Imunológica/imunologia , Feminino , Fluconazol/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Humanos , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-2/administração & dosagem , Interleucina-2/imunologia , Ativação Linfocitária/imunologia , Depleção Linfocítica , Linfócitos do Interstício Tumoral/transplante , Masculino , Melanoma/tratamento farmacológico , Melanoma/imunologia , Pessoa de Meia-Idade , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados , Vidarabina/uso terapêutico
17.
Anticancer Res ; 29(1): 145-54, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19331144

RESUMO

BACKGROUND: Interleukin-2 (IL-2) shows encouraging clinical results in metastatic renal cell carcinoma (RCC) patients, but is limited by substantial toxicity. Cell-based therapy holds a promise, but past attempts in RCC were unsuccessful. Advances in tumor-infiltrating lymphocytes (TIL) generation technology and modified clinical protocols recently yielded a 50% response in refractory melanoma patients. MATERIALS AND METHODS: RCC-derived TIL and tumor cells were processed by current protocols from tumor specimens in a clean laboratory. The expanded TIL were characterized and tested in functional assays. RESULTS: The TIL cultures were efficiently generated and massively expanded. Virtually all the expanded cells were T-cells, but a considerable variability in the CD4/CD8 ratio and a frequent CD4-CD8-phenotype were observed. The TIL exerted cytotoxic or IFNgamma-release activities against autologous targets in major histocompatibility (MHC) class I-restricted and -independent functional patterns. The functional results were superior to former technologies. CONCLUSION: Recent developments in TIL generation technology and clinical patient conditioning protocols indicate that the TIL-based approach for RCC could be revisited.


Assuntos
Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/terapia , Imunoterapia Adotiva/métodos , Neoplasias Renais/imunologia , Neoplasias Renais/terapia , Linfócitos do Interstício Tumoral/imunologia , Relação CD4-CD8 , Carcinoma de Células Renais/patologia , Humanos , Neoplasias Renais/patologia , Estadiamento de Neoplasias , Linfócitos T/imunologia
18.
Immunology ; 126(2): 186-200, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18557789

RESUMO

An efficient immune response against tumours depends on a well-orchestrated function of integrated components, but is finally exerted by effector tumour-infiltrating lymphocytes (TIL). We have previously reported that homophilic CEACAM1 interactions inhibit the specific killing and interferon-gamma (IFN-gamma) release activities of natural killer cells and TIL. In this study a model for the investigation of melanoma cells surviving long coincubation with antigen-specific TIL is reported. It is demonstrated that the surviving melanoma cells increase their surface CEACAM1 expression, which in turn confers enhanced resistance against fresh TIL. Furthermore, it is shown that this is an active process, driven by specific immune recognition and is at least partially mediated by lymphocyte-derived IFN-gamma. Similar results were observed with antigen-restricted TIL, either autologous or allogeneic, as well as with natural killer cells. The enhanced CEACAM1 expression depends, however, on the presence of IFN-gamma and sharply drops within 48 hr. This may be a mechanism that allows tumour cells to transiently develop a more resistant phenotype upon recognition by specific lymphocytes. Therefore, this work substantiates the melanoma-promoting role of CEACAM1 and marks it as an attractive target for novel immunotherapeutic interventions.


Assuntos
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Tolerância Imunológica/imunologia , Melanoma/imunologia , Antígeno 12E7 , Sobrevivência Celular/imunologia , Técnicas de Cocultura , Meios de Cultivo Condicionados , Citotoxicidade Imunológica , Antígeno HLA-A2/metabolismo , Humanos , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas Recombinantes , Células Tumorais Cultivadas , Regulação para Cima/imunologia
19.
J Immunother ; 31(6): 563-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18528298

RESUMO

Adoptive cell transfer represents an important mode of cancer immunotherapy and posttransplant associated viral infections. This technique involves massive volume reduction, as the procedure starts with large-scale ex vivo expansion of lymphocyte cultures (volume up to 60 L), which are harvested at the end of the in vitro process and terminates in a small volume to be infused into the patient. Toward this end, we develop a novel efficient process based on the COBE Spectra apheresis machine usually used for apheresis process. As the COBE Spectra cell separator is easy to use and often available at medical centers, this novel technique is applicable to many transplant and cell processing centers. Our results show a high recovery yield (98%+/-15%) and viability (ranged 79% to 99%) of the large-scale expanded lymphocytes. It preserves sterility of the product and is therefore suitable for immunotherapy treatments.


Assuntos
Imunoterapia Adotiva , Linfócitos/imunologia , Separação Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Humanos , Leucaférese/instrumentação , Masculino
20.
Biochem Biophys Res Commun ; 368(4): 930-6, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18279661

RESUMO

ABC transporters are often found to be inherently expressed in a wide variety of stem cells, where they provide improved protection from toxins. We found a subpopulation of human melanoma cells expressing multidrug-resistance gene product 1 (MDR1). This fraction co-expresses the ABC transporters, ABCB5 and ABCC2 in addition to the stem cell markers, nanog and human telomerase reverse transcriptase (hTERT). The clonogenicity and self-renewal capacity of MDR1(+) melanoma cells were investigated in single cell settings using the limiting dilution assay. We found that the MDR1(+) cells, isolated by FACS sorting, demonstrated a higher self-renewal capacity than the MDR1(-) fraction, a key stem cell feature. Moreover, MDR1(+) cells had higher ability to form spheres in low attachment conditions, a hallmark of cancer. In conclusion, these novel findings imply that the MDR1(+) cells represent melanoma stem cells and thus should be considered as a unique cellular target for future anti-melanoma therapies.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Melanoma/patologia , Células-Tronco Neoplásicas/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Proteínas de Ligação a DNA/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Melanoma/metabolismo , Proteínas de Membrana Transportadoras/biossíntese , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Proteína Homeobox Nanog , Telomerase/biossíntese , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...