Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cancer Immunol Immunother ; 64(8): 941-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25904200

RESUMEN

Human T cells expressing γδ T cell receptor have a potential to show antigen-presenting cell-like phenotype and function upon their activation. However, the mechanisms that underlie the alterations in human γδ T cells remain largely unclear. In this study, we have investigated the molecular characteristics of human γδ T cells related to their acquisition of antigen-presenting capacity in comparison with activated αß T cells. We found that activated γδ but not αß T cells upregulated cell surface expression of a scavenger receptor, CD36, which seemed to be mediated by signaling through mitogen-activated protein kinase and/or NF-κB pathways. Confocal microscopical analysis revealed that activated γδ T cells can phagocytose protein antigens. Activated γδ T cells could induce tumor antigen-specific CD8(+) T cells using both apoptotic and live tumor cells as antigen resources. Furthermore, we detected that C/EBPα, a critical transcription factor for the development of myeloid-lineage cells, is expressed much higher in γδ T cells than in αß T cells. These results unveiled the molecular mechanisms for the elicitation of antigen-presenting functions in γδ T cells and would also help designing new approaches for γδ T cell-mediated human cancer immunotherapy.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Mieloides/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Antígenos CD36/genética , Antígenos CD36/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Línea Celular Tumoral , Linaje de la Célula , Citotoxicidad Inmunológica , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Imidazoles/farmacología , Inmunoterapia/métodos , Activación de Linfocitos , Antígeno MART-1/inmunología , FN-kappa B/antagonistas & inhibidores , Neoplasias/inmunología , Neoplasias/terapia , Fagocitosis , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
2.
Cancer Immunol Res ; 3(1): 26-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25127875

RESUMEN

To understand global effector mechanisms of CTL therapy, we performed microarray gene expression analysis in a murine model using pmel-1 T-cell receptor (TCR) transgenic T cells as effectors and B16 melanoma cells as targets. In addition to upregulation of genes related to antigen presentation and the MHC class I pathway, and cytotoxic effector molecules, cell-cycle-promoting genes were downregulated in the tumor on days 3 and 5 after CTL transfer. To investigate the impact of CTL therapy on the cell cycle of tumor cells in situ, we generated B16 cells expressing a fluorescent ubiquitination-based cell-cycle indicator (B16-fucci) and performed CTL therapy in mice bearing B16-fucci tumors. Three days after CTL transfer, we observed diffuse infiltration of CTLs into the tumor with a large number of tumor cells arrested at the G1 phase of the cell cycle, and the presence of spotty apoptotic or necrotic areas. Thus, tumor growth suppression was largely dependent on G1 cell-cycle arrest rather than killing by CTLs. Neutralizing antibody to IFNγ prevented both tumor growth inhibition and G1 arrest. The mechanism of G1 arrest involved the downregulation of S-phase kinase-associated protein 2 (Skp2) and the accumulation of its target cyclin-dependent kinase inhibitor p27 in the B16-fucci tumor cells. Because tumor-infiltrating CTLs are far fewer in number than the tumor cells, we propose that CTLs predominantly regulate tumor growth via IFNγ-mediated profound cytostatic effects rather than via cytotoxicity. This dominance of G1 arrest over other mechanisms may be widespread but not universal because IFNγ sensitivity varied among tumors.


Asunto(s)
Puntos de Control del Ciclo Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Interferón gamma/inmunología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Fase G1 , Expresión Génica , Humanos , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
Int Immunopharmacol ; 23(2): 499-504, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25284343

RESUMEN

Dendritic cell (DC) vaccine is a promising immunotherapy for cancer due to its ability to induce antigen-specific CTLs efficiently. However, a number of clinical studies have implied insufficient therapeutic benefits with the use of MHC class 1 restricted peptide-pulsed DC vaccine. To enhance the clinical efficacy, we examined combination therapy of DC vaccine pulsed with OVA peptide and intravenous low dose unmodified IL-2 (IL-2 solution) administration against EG7 tumor-bearing mice. Unexpectedly, no additional effects of IL-2 solution were observed on CTL induction and the therapeutic effects of DC vaccine, possibly because of the short half-life of IL-2 in plasma. Therefore, we generated IL-2-encapsulating polymeric micelles (IL-2 micelle), which showed prolonged IL-2 retention in the circulation after intravenous administration compared with IL-2 solution. When mice were treated with OVA peptide-pulsed DCs in combination with IL-2 micelle, OVA-specific CTLs were efficiently induced in the spleen in comparison with DC vaccine combined with IL-2 solution or DC vaccine alone. In addition, combination therapy of DC vaccine and IL-2 micelle against EG7 tumor-bearing mice induced the efficient accumulation of antigen-specific CTLs into the tumor and marked anti-tumor effects. Thus, the administration of IL-2 micelle can significantly enhance DC vaccine efficacy against tumors.


Asunto(s)
Antígenos/metabolismo , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Interleucina-2/farmacología , Neoplasias Experimentales/terapia , Linfocitos T Citotóxicos/fisiología , Animales , Quimioterapia Combinada , Femenino , Regulación Neoplásica de la Expresión Génica , Inmunoterapia , Interleucina-2/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Micelas , Ovalbúmina/genética , Ovalbúmina/metabolismo , Fragmentos de Péptidos , Polímeros , Ratas , Ratas Wistar
4.
Int J Oncol ; 45(5): 1847-56, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25189159

RESUMEN

Specific cellular immunotherapy for cancer requires efficient generation and expansion of cytotoxic T lymphocytes (CTLs) that recognize tumor-associated antigens. However, it is difficult to isolate and expand functionally active T-cells ex vivo. In this study, we investigated the efficacy of a new method to induce expansion of antigen-specific CTLs for adoptive immunotherapy. We used tumor-associated antigen glypican-3 (GPC3)-derived peptide and cytomegalovirus (CMV)-derived peptide as antigens. Treatment of human peripheral blood mononuclear cells (PBMCs) with zoledronate is a method that enables large-scale γδ T-cell expansion. To induce expansion of γδ T cells and antigen-specific CTLs, the PBMCs of healthy volunteers or patients vaccinated with GPC3 peptide were cultured with both peptide and zoledronate for 14 days. The expansion of γδ T cells and peptide-specific CTLs from a few PBMCs using zoledronate yields cell numbers sufficient for adoptive transfer. The rate of increase of GPC3­specific CTLs was approximately 24- to 170,000-fold. These CD8(+) cells, including CTLs, showed GPC3-specific cytotoxicity against SK-Hep-1/hGPC3 and T2 pulsed with GPC3 peptide, but not against SK-Hep-1/vec and T2 pulsed with human immunodeficiency virus peptide. On the other hand, CD8(-) cells, including γδ T cells, showed cytotoxicity against SK-Hep-1/hGPC3 and SK-Hep-1/vec, but did not show GPC3 specificity. Furthermore, adoptive cell transfer of CD8(+) cells, CD8(-) cells, and total cells after expansion significantly inhibited tumor growth in an NOD/SCID mouse model. This study indicates that simultaneous expansion of γδ T cells and peptide-specific CTLs using zoledronate is useful for adoptive immunotherapy.


Asunto(s)
Glipicanos/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Citomegalovirus/inmunología , Difosfonatos/administración & dosificación , Humanos , Imidazoles/administración & dosificación , Leucocitos Mononucleares/inmunología , Ratones , Proteínas de Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Péptidos/inmunología , Ácido Zoledrónico
5.
Anticancer Res ; 34(8): 4601-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25075106

RESUMEN

Several types of immune-cell therapies, such as αß T-cell, γδ T-cell, and dendritic cell (DC) vaccine therapies, are clinically employed for cancer treatment. The safety of immune-cell therapy for the treatment of patients with malignancies should be maintained by continuous assessment of adverse events. In the present study, we surveyed the adverse events associated with immune-cell therapy using large-scale prospective data and analyzed the side-effect profiles. For the assessment of adverse events associated with immune-cell therapy, we evaluated 771 treatment profiles (484 for αß T-cell therapy, 58 for γδ T-cell therapy, 206 for DC vaccine therapy, and 23 for concurrent therapy with αß T-cells and DC vaccines) from 144 patients with various malignancies. For the assessment of fever, fatigue, and itching, each of these adverse events was found to be grade 1 or 2 in most of the treated patients, except for one patient who had grade 3 itching. It was suggested that αß T-cell therapy could elicit a more rapid and direct immune reaction in patients than DC vaccine therapy, as shown by the earlier development of fever and higher incidence rate of fatigue. It was found that grade 1 or 2 reaction at the injection site developed in 10.2% of the patients injected with DC vaccines. Most of the grade 3 non-hematological and hematological adverse events were associated with disease progression or side-effects of chemotherapy, and were not considered to be related to immune-cell therapy. In conclusion, immune-cell therapy, such as αß T-cell, γδ T-cell, or DC vaccine therapy, was well-tolerated for cancer treatment.


Asunto(s)
Vacunas contra el Cáncer/efectos adversos , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/efectos adversos , Neoplasias/terapia , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Receptores de Antígenos de Linfocitos T alfa-beta , Receptores de Antígenos de Linfocitos T gamma-delta
6.
Int Immunopharmacol ; 18(1): 90-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24269583

RESUMEN

Recent progress has been made in understanding the mechanisms of antitumor immune responses, which may further clarify the immune status of cancer patients. In this study, we performed a detailed evaluation of the immunological status of 47 patients with advanced solid cancer, who had received no immunosuppressive treatment, and compared the results with 32 healthy subjects. Flow-cytometry data for peripheral blood were obtained using 19 monoclonal antibodies against various cell surface and intracellular molecules. Absolute numbers of T cells, several T cell subsets, B cells, and NK cells were significantly decreased in patients compared with healthy subjects. The percentage of CD27(+)CD45RA(+) T cells was lower and that of CD27(-)CD45RA(-) T cells was higher in patients compared with controls. Regulatory and type 2 helper T cells were elevated in patients relative to healthy subjects. The percentage of perforin(+) NK cells was significantly lower in patients than in controls. These results suggest a dysfunctional anti-tumor immune response in cancer patients. Furthermore, peripheral blood from 26 of 47 cancer patients was analyzed after adoptive T cell immunotherapy (ATI). ATI increased the number of T cell subsets, but not B and NK cells. The number and percentage of regulatory T cells decreased significantly. These results suggest that ATI can restore impaired and imbalanced T cell immune status.


Asunto(s)
Linfocitos B/inmunología , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Células , Células Cultivadas , Citocinas/metabolismo , Femenino , Estado de Salud , Homeostasis , Humanos , Inmunidad Celular , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/diagnóstico , Resultado del Tratamiento
7.
Int J Cancer ; 134(8): 1810-22, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24150772

RESUMEN

Complex interactions among multiple cell types contribute to the immunosuppressive milieu of the tumor microenvironment. Using a murine model of adoptive T-cell immunotherapy (ACT) for B16 melanoma, we investigated the impact of tumor infiltrating cells on this complex regulatory network in the tumor. Transgenic pmel-1-specific cytotoxic T lymphocytes (CTLs) were injected intravenously into tumor-bearing mice and could be detected in the tumor as early as on day 1, peaking on day 3. They produced IFN-γ, exerted anti-tumor activity and inhibited tumor growth. However, CTL infiltration into the tumor was accompanied by the accumulation of large numbers of cells, the majority of which were CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs). Notably, CD11b(+) Gr1(int) Ly6G(-) Ly6C(+) monocytic MDSCs outnumbered the CTLs by day 5. They produced nitric oxide, arginase I and reactive oxygen species, and inhibited the proliferation of antigen-specific CD8(+) T cells. The anti-tumor activity of the adoptively-transferred CTLs and the accumulation of MDSCs both depended on IFN-γ production on recognition of tumor antigens by the former. In CCR2(-/-) mice, monocytic MDSCs did not accumulate in the tumor, and inhibition of tumor growth by ACT was improved. Thus, ACT triggered counter-regulatory immunosuppressive mechanism via recruitment of MDSCs. Our results suggest that strategies to regulate the treatment-induced recruitment of these MDSCs would improve the efficacy of immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Interferón gamma/metabolismo , Melanoma Experimental/inmunología , Células Madre Neoplásicas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Arginasa/biosíntesis , Antígeno CD11b/metabolismo , Proliferación Celular , Células Cultivadas , Células Dendríticas/inmunología , Terapia de Inmunosupresión , Interferón gamma/biosíntesis , Masculino , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Óxido Nítrico/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR2/deficiencia , Receptores CCR2/genética , Linfocitos T Citotóxicos/metabolismo , Microambiente Tumoral
8.
Oncoimmunology ; 2(9): e25636, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24319636

RESUMEN

The addition of zoledronate to tumor-associated antigen (TAA)-loaded dendritic cells (DCs) promotes the activation of interferon γ-secreting Vγ9 γδ T cells, in turn eliciting TAA-specific CD8+ T-cell responses. Immunological responses induced by zoledronate-pulsed DC-based vaccines have been associated with therapeutic effects in clinical trials.

9.
Anticancer Res ; 33(7): 2971-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23780988

RESUMEN

Dendritic cell (DC)-based vaccines with the use of various antigen loading methods have been developed for cancer immunotherapy. Electroporation (EP) of a whole tumor cell lysate into DCs was previously found to be more potent for eliciting antigen-specific CD8 + T-cells compared to co-incubation of tumor cell lysates with DCs in vitro. In the present report, we studied the feasibility, safety and antitumor effect in the clinical use of an EP-DC vaccine for the immunotherapy of various types of human solid tumors. We successfully prepared an autologous tumor lysate-loaded EP-DC vaccine with high cell viability by the closed-flow electroporation system. In the phase I clinical trial, mild adverse events associated with the EP-DC vaccine were found during the treatment of advanced or recurrent cancer, or during the adjuvant therapy of some types of cancer; no autoimmune responses were observed after treatment with the autologous tumor lysate-loaded EP-DC vaccines. For the antitumor effect of the EP-DC vaccine against the 41 various types of solid tumor, the overall response rate [complete remission (CR) + partial response (PR)] was 4.9% (2/41) and the clinical benefit rate [CR+ PR + long stable disease (SD)] was 31.7% (13/41). Furthermore, the delayed-type hypersensitivity (DTH) reactivity was positive in most cases of long SD and the positive rate of DTH was 91.7% (11/12) for the patients with clinical benefit. In conclusion, the safety and feasibility of the EP-DC vaccine with autologous tumor lysates were confirmed, and it was found that the antitumor effect might be associated with the immunological response induced by the EP-DC vaccine for cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Electroporación , Hipersensibilidad Tardía/etiología , Inmunoterapia , Neoplasias/terapia , Adulto , Anciano , Antígenos de Neoplasias/metabolismo , Células Dendríticas/metabolismo , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Neoplasias/metabolismo , Pronóstico , Linfocitos T Citotóxicos/inmunología
10.
Int Immunopharmacol ; 15(3): 488-97, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23474736

RESUMEN

Recent commercial approval of cancer vaccine, demonstrating statistically significant improvement in overall survival of prostate cancer patients has spurred renewed interest in active immunotherapies; specifically, strategies that lead to enhanced biological activity and robust efficacy for dendritic cell vaccines. A simple, widely used approach to generating multivalent cancer vaccines is to load tumor whole cell lysates into dendritic cells (DCs). Current DC vaccine manufacturing processes require co-incubation of tumor lysate antigens with immature DCs and their subsequent maturation. We compared electroloading of tumor cell lysates directly into mature DCs with the traditional method of lysate co-incubation with immature DCs. Electroloaded mature DCs were more potent in vitro, as judged by their ability to elicit significantly (p < 0.05) greater expansion of peptide antigen-specific CD8(+) T cells, than either lysate-electroloaded immature DCs or lysate-co-incubated immature DCs, both of which must be subsequently matured. Expanded CD8(+) T cells were functional as judged by their ability to produce IFN-γ upon antigen-specific re-stimulation. The electroloading technology used herein is an automated, scalable, functionally closed cGMP-compliant manufacturing technology supported by a Master File at CBER, FDA and represents an opportunity for translation of enhanced potency DC vaccines at clinical/commercial scale.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/metabolismo , Electroporación/métodos , Inmunoterapia Adoptiva/métodos , Melanoma/inmunología , Linfocitos T Citotóxicos/inmunología , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígenos CD8/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/citología , Estudios de Factibilidad , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Melanoma/terapia
12.
Cancer Res ; 68(10): 3941-9, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18483280

RESUMEN

Targeting dendritic cells in vivo by transcutaneous peptide immunization (TCI) represents an efficient immunization strategy to induce tumor-specific CTL because it reflects the physiologic conditions occurring during pathogen infection. Here we show that including a Th1 peptide in TCI can activate preexisting memory Th1 (mTh1) responses and thereby enhance the CTL response. For this purpose, peptide-25, a major Th1 epitope of Ag85B from Mycobacterium tuberculosis, was selected. We adoptively transferred peptide-25-specific mTh1 cells and hgp100-specific naive CTL (pmel-1 TCR transgenic) into C57BL/6 mice. Subsequently, mice were transcutaneously immunized with CTL peptide (hgp100) and Th1 peptide (peptide-25). Five days after TCI, the frequency and function of pmel-1 cells was monitored by intracellular IFN-gamma staining, ELISPOT, and in vivo cytotoxicity assays. TCI efficiently expanded hgp100-specific, IFN-gamma-producing, strongly cytotoxic CD8(+) T cells. Concurrent activation of mTh1 cells by peptide-25 induced a 1.5-fold increase in the number of hgp100-specific CTL with enhanced effector functions. Furthermore, TCI elicited not only prophylactic but also therapeutic antitumor responses that were augmented by peptide-25. These results show that TCI facilitates peptide-specific activation of CD4(+) T cells, responsible for the augmenting effect of peptide-25 on the hgp100-specific CTL response. Because a significant proportion of the Japanese population has been vaccinated with Bacillus Calmette-Guerin, they are likely to possess Ag85B- or peptide-25-specific mTh1 cells. Therefore, concomitant activation of Ag85B- or peptide-25-specific mTh1 cells together with tumor-specific CTL by TCI might augment antitumor immune responses in a sizeable fraction of patients.


Asunto(s)
Péptidos/química , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/citología , Células TH1/inmunología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Epítopos/química , Memoria Inmunológica , Interferón gamma/metabolismo , Leucocitos Mononucleares/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células TH1/citología
13.
Biochem Biophys Res Commun ; 371(2): 242-6, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18423376

RESUMEN

Dendritic cells (DCs) transfected with mRNA encoding tumor-associated antigens (TAAs) can induce tumor-specific T-cell responses. To potentiate this, we transfected mature DCs (mDCs) with mRNA encoding TAA targeted to the proteasome. DCs were generated from bone marrow cells by culture with 20 ng/ml GM-CSF and maturation with 1 microg/ml LPS. These mDCs were then electroporated with 10 microg of mRNA. Antigen presentation after electroporation with in vitro transcribed mRNA was compared with mRNA from a construct of the TAA preceded by ubiquitin. Proteasomal targeting of mRNA encoding cotranslationally ubiquitinated antigen was found to enhance intracellular degradation of target protein, and result in more efficient priming and expansion of TAA-specific CD8(+) T-cells. We therefore suggest that RNA-transfected DC vaccine efficacy could be improved by the use of mRNA targeted to the proteasome.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Linfocitos T Citotóxicos/inmunología , Ubiquitinación , Animales , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Técnicas de Cultivo de Célula , Células Dendríticas/efectos de los fármacos , Electroporación , Antígeno MART-1 , Ratones , Ratones Endogámicos , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Ubiquitinación/genética
14.
Bioorg Med Chem Lett ; 14(10): 2597-601, 2004 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15109660

RESUMEN

Two analogues possessing steric hindered substituents on C-15 of arenastatin A (1), a potent cytotoxic spongean depsipeptide, were synthesized and shown to enhance stability in mouse serum. Notably, 15-tert-butylanalogue (6) with higher cytotoxicity exhibited in vivo anti-tumor activity through iv administration different from 1. Additionally, conformation analysis among the two analogues and arenastatin A (1) indicated that the torsion angle from C-14 to C-20 is a conclusive factor for the potent cytotoxicity of 1.


Asunto(s)
Antineoplásicos/síntesis química , Depsipéptidos/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Citotoxinas , Depsipéptidos/síntesis química , Depsipéptidos/uso terapéutico , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Ratones , Conformación Molecular , Poríferos/química , Relación Estructura-Actividad , Resultado del Tratamiento
15.
Neuromuscul Disord ; 13(10): 796-803, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14678802

RESUMEN

Alpha-dystroglycan is a cell surface peripheral membrane protein which binds to the extracellular matrix, while beta-dystroglycan is a type I integral membrane protein which anchors alpha-dystroglycan to the cell membrane via the N-terminal extracellular domain. The complex composed of alpha- and beta-dystroglycan is called the dystroglycan complex. Although defects of the dystroglycan gene have not been identified as the primary causes of hereditary diseases in humans, secondary but significant abnormalities of the dystroglycan complex have been revealed in severe muscular dystrophies, including sarcoglycanopathy (LGMD2C, D, E and F). In this study, we investigated proteolytic processing of beta-dystroglycan and its effect on the extracellular matrix-cell membrane linkage in cardiomyopathic hamsters, the model animals of LGMD2F. Compared to normal controls, proteolytic processing of beta-dystroglycan was activated in the skeletal, cardiac and smooth muscles of cardiomyopathic hamsters and this resulted in the partial disruption of the dystroglycan complex in these tissues. These phenomena were observed from the early phase of muscle degeneration process. Our results suggest that proteolytic processing of beta-dystroglycan disrupts the extracellular matrix-cell membrane linkage via the dystroglycan complex and this may play a role in the molecular pathogenesis of muscle degeneration in cardiomyopathic hamsters.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas del Citoesqueleto/metabolismo , Glicoproteínas de Membrana/metabolismo , Músculos/metabolismo , Animales , Encéfalo/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Línea Celular , Membrana Celular/metabolismo , Cricetinae , Dimerización , Modelos Animales de Enfermedad , Distroglicanos , Matriz Extracelular/metabolismo , Sustancias Macromoleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Liso/metabolismo , Músculo Liso/patología , Músculos/patología , Músculos/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Péptido Hidrolasas/metabolismo , Ratas
16.
Gan To Kagaku Ryoho ; 29(11): 1943-9, 2002 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-12465394

RESUMEN

The antitumor activity of a combination of paclitaxel (TXL) followed by nedaplatin (NDP) against SK-OV-3 human ovarian cancer was evaluated. We also compared the antitumor activity of TXL plus NDP with that of TXL plus carboplatin (CBDCA) or TXL plus cisplatin (CDDP). TXL was injected i.v. daily for four days and either NDP, CBDCA or CDDP was injected i.v. once after the TXL treatment, into tumor-bearing mice. The sequential administration of TXL prior to NDP resulted in enhanced inhibition of tumor growth in comparison with either TXL or NDP monotherapy. The combination in TXL plus NDP was synergistic and superior to that of TXL plus CDDP or TXL plus CBDCA therapy. Histological tests demonstrated that the fraction of BrdU-incorporated cells in tumor tissue was significantly inhibited by the combination of TXL with NDP. These results demonstrated the antitumor efficacy of TXL with NDP against human ovarian cancer and suggest the clinical effectiveness of combination of TXL with NDP.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Compuestos Organoplatinos/administración & dosificación , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación
17.
Clin Exp Metastasis ; 19(6): 519-26, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12405289

RESUMEN

The anti-metastatic efficacy of MMI-166, which is a selective matrix metalloproteinase (MMP) inhibitor, in combination with CPT-11 was examined using two metastasis models of human gastrointestinal cancer cells. In the liver metastasis model, C-IH human colon cancer cells were injected into the spleen of athymic BALB/c nude mice. Daily oral (p.o.) dosing of MMI-166 at 200 mg/kg starting 1 day after tumor inoculation led to a significantly prolonged survival effect by inhibiting liver metastasis of C-1H tumor cells. CPT-11 (5 or 20 mg/kg) was administered intraperitoneally (i.p.) three times on day 3, day 7 and day 11 and also improved the survival of tumor-inoculated mice compared with the vehicle control. When MMI-166 was combined with CPT-11, the anti-metastatic efficacy was significantly augmented. Moreover, long tumor-free survival was noted in two of eight mice that were given the combination therapy but not either MMI-166 or CPT-11 monotherapy. In the peritoneal dissemination model, TMK-1 human gastric cancer cells were injected i.p. into nude mice. While both MMI-166, administered daily p.o. from day 1 at 200 mg/kg, and CPT-11, administered intravenously (i.v.) three times, inhibited the tumor dissemination and growth, the combination therapy of MMI-166 plus CPT-11 showed a greater inhibitory effect than each monotherapy. A hematotoxicity study demonstrated that CPT-11 alone significantly decreased the number of white blood cells (WBC) and bone marrow cells (BMC) in the mice during treatment, while the daily administration of MMI-166 alone had no such effect. More importantly, the combination therapy of MMI-166 with CPT-11 did not augment the hematotoxicity caused by CPT-11. An in vitro cytotoxicity study showed that MMI-166 itself neither has direct cytotoxicity in C-1H and TMK-1 tumor cells, nor does it augment the cytotoxicity of SN-38, an active form of CPT-11. The findings indicate that the augmented anti-metastatic efficacy in combination treatment was not simply due to the augmentation of direct cytotoxic activity, but was rather an additive or synergistic effect of anti-metastatic activities with different mechanisms. In conclusion, we demonstrated that the anti-metastatic efficacy against C-1H colon cancer and TMK-1 gastric cancer were augmented by the combination therapy of MMI-166, an orally active MMP inhibitor, with CPT-11. However, the hematotoxicity caused by CPT-11 was not augmented in the combination with MMI-166. Thus, the combination therapy of MMI-166 and CPT-11 exhibited potent anti-metastatic efficacy without increased hematotoxicity. These results point to the clinical advantage of using MMI-166 in combination with CPT-11.


Asunto(s)
Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Neoplasias del Colon/patología , Neoplasias Hepáticas/secundario , Inhibidores de la Metaloproteinasa de la Matriz , Metástasis de la Neoplasia/prevención & control , Sulfonamidas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Irinotecán , Neoplasias Hepáticas/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia/patología , Trasplante Heterólogo
18.
Cancer Lett ; 182(1): 61-8, 2002 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-12175524

RESUMEN

The antitumor efficacy of the combination of nedaplatin (NDP) with gemcitabine (GEM) was evaluated against Ma44/GEM, a GEM-refractory subline of Ma44 human lung cancer, which was established by serial in vitro passage of Ma44 cells in the presence of GEM.Ma44/GEM showed less sensitivity to GEM and cytosine arabinoside with resistance factors of 7.7 and 8.3, respectively, but not to Taxol, Irinotecan, Mitomycin C and NDP. Flow cytometry analysis demonstrated that membrane transporter molecules such as multidrug-resistant, multidrug-resistant related protein or lung resistant protein were not induced in Ma44/GEM cells. In vivo experiments confirmed the less sensitivity of Ma44/GEM to GEM. The resistant factor of Ma44/GEM to GEM in vivo was estimated to be 6.7 in terms of ED(50).MA44/GEM-implanted athymic mice were treated with GEM i.v. once followed by i.v. injection of NDP at an interval of approximately 30 min. The mice were treated again with GEM after 3 or 4 days. The combined dosing of NDP with GEM resulted in synergistically enhanced inhibition of tumor growth against Ma44/GEM. The antitumor efficacy of the combination of NDP and GEM was superior to the best effect of either monotherapy. These results demonstrate the effectiveness of the combination of NDP with GEM against the GEM-refractory human lung cancer model.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Desoxicitidina/análogos & derivados , Desoxicitidina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Compuestos Organoplatinos/administración & dosificación , Partículas Ribonucleoproteicas en Bóveda , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/análisis , Animales , Citarabina/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo , Células Tumorales Cultivadas , Gemcitabina
19.
J Control Release ; 82(2-3): 183-7, 2002 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-12175736

RESUMEN

Recombinant interleukin-2 (IL-2) was strongly and almost completely adsorbed onto small hydrophobic liposomes under optimal conditions (liposome: DSPC-DSPG; molar ratio, 10:1; 30-50 nm in size, ratio of IL-2 to liposome: 4.0 JRU/nmol lipid). This liposomal IL-2 improved the distribution of IL-2 after intravenous administration as reported, previously. Liposomal IL-2 (300-10000 JRU/mouse per day) was significantly more effective than free IL-2 alone for inhibiting against the experimental metastases of M5076 in mice. The inhibitory effect of liposomal IL-2 was greatest in the liver. The ED(50) of liposomal IL-2 and that of free IL-2 in the liver were 1640 and 12500 JRU/mouse per day, respectively. This simple preparation (mixture) using IL-2 and liposome suspension is expected to have potential for increasing therapeutic efficacy against hepatic metastases.


Asunto(s)
Antineoplásicos/administración & dosificación , Interleucina-2/administración & dosificación , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/secundario , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteínas Recombinantes/administración & dosificación , Animales , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Composición de Medicamentos , Femenino , Inyecciones Intravenosas , Interleucina-2/uso terapéutico , Liposomas , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos , Proteínas Recombinantes/uso terapéutico
20.
J Neuroimmunol ; 127(1-2): 134-8, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12044984

RESUMEN

Matrix metalloproteinases (MMPs) have been reported to be involved in various inflammatory disorders. Previous studies revealed that MMP-2 and MMP-9 might play important roles in the breakdown of the blood-brain barrier (BBB) in the central nervous system (CNS) of patients with HTLV-I-associated myelopathy (HAM)/tropical spastic paraparesis (TSP). N-Biphenyl sulfonyl-phenylalanine hydroxamic acid (BPHA) selectively inhibits MMP-2, -9 and -14, but not MMP-1, -3 and -7. In the present study, we examined whether or not the selective MMP inhibitor BPHA could inhibit the heightened migrating activity of CD4+ T cells in HAM/TSP patients. The migration assay using an invasion chamber showed that migration of CD4+ T cells in HAM/TSP patients was inhibited by 25 microM BPHA. In addition, the inhibitory ratio of migrating CD4+ lymphocytes was higher in HAM patients compared to normal controls. These results suggest that the selective MMP inhibitor BPHA has therapeutic potential for HAM/TSP.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácidos Hidroxámicos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Paraparesia Espástica Tropical/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Materiales Biocompatibles , Linfocitos T CD4-Positivos/citología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Colágeno , Combinación de Medicamentos , Femenino , Humanos , Laminina , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Persona de Mediana Edad , Paraparesia Espástica Tropical/tratamiento farmacológico , Paraparesia Espástica Tropical/enzimología , Proteoglicanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...