Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Blood ; 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38518105

RESUMEN

Acute lymphoblastic leukemia (ALL) arises from the uncontrolled proliferation of precursor B or T cells (BCP- or T-ALL). Current treatment protocols obtain high cure rates in children but are based on toxic polychemotherapy. Novel therapies are urgently needed, especially in relapsed/refractory (r/r) disease, high-risk leukemias and T-ALL, where immunotherapy approaches remain scarce. While the Interleukin-7 receptor (IL-7R) plays a pivotal role in ALL development, no IL-7R-targeting immunotherapy has yet reached clinical application in ALL. The IL-7Rα chain (CD127)-targeting IgG4 antibody Lusvertikimab (formerly OSE-127) is a full antagonist of the IL-7R pathway showing a good safety profile in healthy volunteers. Here, we show that ~85% of ALL cases express surface CD127. We demonstrate significant in vivo efficacy of Lusvertikimab immunotherapy in a heterogeneous cohort of BCP- and T-ALL patient-derived xenografts (PDX) in minimal residual disease (MRD) and overt leukemia models, including r/r and high-risk leukemias. Importantly, Lusvertikimab was particularly effective when combined with polychemotherapy in a phase 2-like PDX study with CD127high samples leading to MRD-negativity in >50% of mice treated with combination therapy. Mechanistically, Lusvertikimab targeted ALL cells via a dual mode of action comprising direct IL-7R antagonistic activity and induction of macrophage-mediated antibody-dependent cellular phagocytosis (ADCP). Lusvertikimab-mediated in vitro ADCP levels significantly correlated with CD127 expression levels and the reduction of leukemia burden upon treatment of PDX animals in vivo. Altogether, through its dual mode of action and good safety profile, Lusvertikimab may represent a novel immunotherapy option for any CD127-positive ALL, particularly in combination with standard-of-care polychemotherapy.

2.
Cancers (Basel) ; 14(22)2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36428719

RESUMEN

Despite advances in clinical management, osteosarcoma and Ewing sarcoma, the two most frequent malignant primary bone tumors at pediatric age, still have a poor prognosis for high-risk patients (i.e., relapsed or metastatic disease). Triggering a TRAIL pro-apoptotic pathway represents a promising therapeutic approach, but previous studies have described resistance mechanisms that could explain the declining interest of such an approach in clinical trials. In this study, eight relevant human cell lines were used to represent the heterogeneity of the response to the TRAIL pro-apoptotic effect in pediatric bone tumors and two cell-derived xenograft models were developed, originating from a sensitive and a resistant cell line. The DR5 agonist antibody AMG655 (Conatumumab) was selected as an example of TRAIL-based therapy. In both TRAIL-sensitive and TRAIL-resistant cell lines, two signaling pathways were activated following AMG655 treatment, the canonical extrinsic apoptotic pathway and a non-apoptotic pathway, involving the recruitment of RIPK1 on the DR5 protein complex, activating both pro-survival and pro-proliferative effectors. However, the resulting balance of these two pathways was different, leading to apoptosis only in sensitive cells. In vivo, AMG655 treatment reduced tumor development of the sensitive model but accelerated tumor growth of the resistant one. We proposed two independent strategies to overcome this issue: (1) a proof-of-concept targeting of RIPK1 by shRNA approach and (2) the use of a novel highly-potent TRAIL-receptor agonist; both shifting the balance in favor of apoptosis. These observations are paving the way to resurrect TRAIL-based therapies in pediatric bone tumors to help predict the response to treatment, and propose a relevant adjuvant strategy for future therapeutic development.

3.
Bioconjug Chem ; 30(6): 1665-1676, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31045351

RESUMEN

To reply to as yet unmet medical needs to treat osteosarcoma, a form of primary bone cancer, we conceived the 12b80 compound by covalently conjugating antineoplastic compound doxorubicin to a bone targeting hydroxybisphosphonate vector and turned it into a prodrug through a custom linker designed to specifically trigger doxorubicin release in acidic bone tumor microenvironment. Synthesis of 12b80 was thoroughly optimized to be produced at gram scale. 12b80 was evaluated in vitro for high bone support affinity, specific release of doxorubicin in acidic condition, lower cytotoxicity, and cellular uptake of the prodrug. In vivo in rodents, 12b80 displayed rapid and sustained targeting of bone tissue and tumor-associated heterotopic bone and permitted a higher doxorubicin payload in tumor bone environment compared to nonvectorized doxorubicin. Consequently, 12b80 showed much lower toxicity compared to doxorubicin, promoted strong antitumor effects on rodent orthotopic osteosarcoma, displayed a dose-response therapeutic effect, and was more potent than doxorubicin/zoledronate combination.


Asunto(s)
Antibióticos Antineoplásicos/química , Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/química , Doxorrubicina/análogos & derivados , Osteosarcoma/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/síntesis química , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapéutico , Neoplasias Óseas/patología , Línea Celular Tumoral , Técnicas de Química Sintética , Difosfonatos/síntesis química , Difosfonatos/farmacocinética , Difosfonatos/uso terapéutico , Doxorrubicina/síntesis química , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Femenino , Ratones Desnudos , Osteosarcoma/patología , Ratas
4.
Cytotherapy ; 20(8): 1037-1045, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30093324

RESUMEN

BACKGROUND: Osteosarcoma (OS) is the most frequent pediatric malignant bone tumor. OS patients have not seen any major therapeutic progress in the last 30 years, in particular in the case of metastatic disease, which requires new therapeutic strategies. The pro-apoptotic cytokine Tumor necrosis factor (TNF)-Related Apoptosis Inducing Ligand (TRAIL) can selectively kill tumor cells while sparing normal cells, making it a promising therapeutic tool in several types of cancer. However, many OS cell lines appear resistant to recombinant human (rh)TRAIL-induced apoptosis. We, therefore, hypothesized that TRAIL presentation at the membrane level of carrier cells might overcome this resistance and trigger apoptosis. METHODS: To address this, human adipose mesenchymal stromal cells (MSCs) transfected in a stable manner to express membrane-bound full-length human TRAIL (mbTRAIL) were co-cultured with several human OS cell lines. RESULTS: This induced apoptosis by cell-to-cell contact even in cell lines initially resistant to rhTRAIL. In contrast, mbTRAIL delivered by MSCs was not able to counteract tumor progression in this OS orthotopic model. DISCUSSION: This was partly due to the fact that MSCs showed a potential to support tumor development. Moreover, the expression of mbTRAIL did not show caspase activation in adjacent tumor cells.


Asunto(s)
Apoptosis , Neoplasias Óseas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osteosarcoma/terapia , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Traslado Adoptivo/métodos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Células Cultivadas , Terapia Genética/métodos , Humanos , Técnicas In Vitro , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Osteosarcoma/genética , Osteosarcoma/patología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Int J Cancer ; 139(12): 2802-2811, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27558972

RESUMEN

Ewing sarcoma (EWS) is the second most frequent pediatric malignant bone tumor. EWS patients have not seen any major therapeutic progress in the last 30 years, in particular in the case of metastatic disease, which requires new therapeutic strategies. The pro-apoptotic cytokine TNF-Related Apoptosis Inducing Ligand (TRAIL) can selectively kill tumor cells while sparing normal cells, making it a promising therapeutic tool in several types of cancer. However, certain EWS cell lines appear resistant to recombinant human (rh) TRAIL-induced apoptosis. We therefore hypothesized that a TRAIL presentation at the surface of the carrier cells might overcome this resistance and trigger apoptosis. For this purpose, human adipose mesenchymal stromal/stem cells (MSC) transfected in a stable manner to express full-length human TRAIL were co-cultured with several human EWS cell lines, inducing apoptosis by cell-to-cell contact even in cell lines initially resistant to rhTRAIL or AMG655, an antibody agonist to the death receptor, DR5. In vivo, TRAIL delivered by MSCs was able to counteract tumor progression in two orthotopic models of Ewing sarcoma, associated with caspase activation, indicating that a cell-based delivery of a potent apoptosis-inducing factor could be relevant in EWS.


Asunto(s)
Transformación Celular Neoplásica/genética , Células Madre Mesenquimatosas/metabolismo , Sarcoma de Ewing/etiología , Sarcoma de Ewing/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Animales , Apoptosis/genética , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Xenoinjertos , Humanos , Ratones , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/patología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción Genética
6.
Am J Cancer Res ; 6(3): 677-89, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27152244

RESUMEN

Osteosarcoma, the most frequent malignant primary bone tumor in pediatric patients is characterized by osteolysis promoting tumor growth. Lung metastasis is the major bad prognosis factor of this disease. Zoledronic Acid (ZA), a potent inhibitor of bone resorption is currently evaluated in phase III randomized studies in Europe for the treatment of osteosarcoma and Ewing sarcoma. The beneficial effect of the liposomal form of Muramyl-TriPeptide-Phosphatidyl Ethanolamine (L-mifamurtide, MEPACT®), an activator of macrophage populations has been demonstrated to eradicate lung metastatic foci in osteosarcoma. The objective of this study was to evaluate the potential therapeutic benefit and the safety of the ZA and L-mifamurtide combination in preclinical models of osteosarcoma, as a prerequisite before translation to patients. The effects of ZA (100 µg/kg) and L-mifamurtide (1 mg/kg) were investigated in vivo in xenogeneic and syngeneic mice models of osteosarcoma, at clinical (tumor proliferation, spontaneous lung metastases development), radiological (bone microarchitecture by microCT analysis), biological and histological levels. No interference between the two drugs could be observed on ZA-induced bone protection and on L-mifamurtide-induced inhibition of lung metastasis development. Unexpectedly, ZA and L-mifamurtide association induced an additional and in some cases synergistic inhibition of primary tumor progression. L-mifamurtide has no effect on tumor proliferation in vitro or in vivo, and macrophage population was not affected at the tumor site whatever the treatment. This study evidenced for the first time a significant inhibition of primary osteosarcoma progression when both drugs are combined. This result constitutes a first proof-of-principle for clinical application in osteosarcoma patients.

7.
Clin Cancer Res ; 20(19): 5097-112, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25107916

RESUMEN

PURPOSE: Osteosarcoma is the main malignant primary bone tumor in children and adolescents for whom the prognosis remains poor, especially when metastasis is present at diagnosis. Because transforming growth factor-ß (TGFß) has been shown to promote metastasis in many solid tumors, we investigated the effect of the natural TGFß/Smad signaling inhibitor Smad7 and the TßRI inhibitor SD-208 on osteosarcoma behavior. EXPERIMENTAL DESIGN: By using a mouse model of osteosarcoma induced by paratibial injection of cells, we assessed the impact of Smad7 overexpression or SD-208 on tumor growth, tumor microenvironment, bone remodeling, and metastasis development. RESULTS: First, we demonstrated that TGFß levels are higher in serum samples from patients with osteosarcoma compared with healthy volunteers and that TGFß/Smad3 signaling pathway is activated in clinical samples. Second, we showed that Smad7 slows the growth of the primary tumor and increases mice survival. We furthermore demonstrated that Smad7 expression does not affect in vitro osteosarcoma cell proliferation but affects the microarchitectural parameters of bone. In addition, Smad7-osteosarcoma bone tumors expressed lower levels of osteolytic factors such as RANKL, suggesting that Smad7 overexpression affects the "vicious cycle" established between tumor cells and bone cells by its ability to decrease osteoclast activity. Finally, we showed that Smad7 overexpression in osteosarcoma cells and the treatment of mice with SD208 inhibit the development of lung metastasis. CONCLUSION: Taken together, these results demonstrate that the inhibition of the TGFß/Smad signaling pathway may be a promising therapeutic strategy against tumor progression of osteosarcoma, specifically against the development of lung metastasis.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/patología , Expresión Génica , Neoplasias Pulmonares/secundario , Osteosarcoma/genética , Osteosarcoma/patología , Proteína smad7/genética , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/mortalidad , Resorción Ósea/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/mortalidad , Pteridinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/sangre , Carga Tumoral/genética
8.
Eur J Cancer ; 49(9): 2253-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23490646

RESUMEN

BACKGROUND: Osteosarcoma is the most common primary malignant bone tumour in children and adolescents for whom the prognosis remains unfavourable despite treatment protocols that combine chemotherapy and surgery. Metalloproteinases decisively contribute to cancer development and promotion by regulating cell growth, angiogenesis or inflammation. However, their role in osteosarcoma remains still unknown. METHODS: A screening of a large panel of metalloproteinases and their inhibitors, carried out in osteolytic (K7M2 and POS-1) or osteoblastic (MOS-J) mouse osteosarcoma models, shows that a member of a family of cell surface metallopeptidases, A Disintegrin And Metalloproteinase 12 (ADAM12), is highly expressed in the K7M2 and POS-1 cell lines and weakly expressed in the MOS-J cell line. To investigate whether ADAM12, involved in several pathologic conditions characterised by abnormal cell growth, plays a role in osteosarcoma tumour growth, ADAM12 was overexpressed in MOS-J and downregulated in K7M2 cells. RESULTS: In vivo experiments demonstrated that ADAM12 favours tumour growth, leading to a significant modification in animal survival. In vitro assays showed that ADAM12 knockdown in K7M2 cells slows cell proliferation. In addition, the study of microarchitectural parameters, assessed by micro-computed tomography (CT) analysis, showed that ADAM12 favours bone osteolysis, as demonstrated both in an ADAM12 overexpressing (MOS-J) and a knockdown (K7M2) model. Histological analysis showed that ADAM12 inhibited osteoblast activity and therefore enhanced bone resorption. CONCLUSIONS: Our study demonstrates that ADAM12 expression not only favours tumour growth but also associates enhanced osteolysis with a significant reduction in animal survival, suggesting that ADAM12 could be a new therapeutic target in osteosarcoma.


Asunto(s)
Proteínas ADAM/fisiología , Neoplasias Óseas/patología , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Osteólisis/patología , Osteosarcoma/patología , Proteínas ADAM/metabolismo , Proteína ADAM12 , Adolescente , Adulto , Animales , Neoplasias Óseas/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Osteólisis/metabolismo , Osteosarcoma/metabolismo , Transfección , Células Tumorales Cultivadas , Adulto Joven
9.
PLoS One ; 5(12): e14398, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21200436

RESUMEN

BACKGROUND: Changes in imprinted gene dosage in the placenta may compromise the prenatal control of nutritional resources. Indeed monoallelic behaviour and sensitivity to changes in regional epigenetic state render imprinted genes both vulnerable and adaptable. METHODS AND FINDINGS: We investigated whether a high-fat diet (HFD) during pregnancy modified the expression of imprinted genes and local and global DNA methylation patterns in the placenta. Pregnant mice were fed a HFD or a control diet (CD) during the first 15 days of gestation. We compared gene expression patterns in total placenta homogenates, for male and female offspring, by the RT-qPCR analysis of 20 imprinted genes. Sexual dimorphism and sensitivity to diet were observed for nine genes from four clusters on chromosomes 6, 7, 12 and 17. As assessed by in situ hybridization, these changes were not due to variation in the proportions of the placental layers. Bisulphite-sequencing analysis of 30 CpGs within the differentially methylated region (DMR) of the chromosome 17 cluster revealed sex- and diet-specific differential methylation of individual CpGs in two conspicuous subregions. Bioinformatic analysis suggested that these differentially methylated CpGs might lie within recognition elements or binding sites for transcription factors or factors involved in chromatin remodelling. Placental global DNA methylation, as assessed by the LUMA technique, was also sexually dimorphic on the CD, with lower methylation levels in male than in female placentae. The HFD led to global DNA hypomethylation only in female placenta. Bisulphite pyrosequencing showed that neither B1 nor LINE repetitive elements could account for these differences in DNA methylation. CONCLUSIONS: A HFD during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes important in the control of many cellular, metabolic and physiological functions potentially involved in adaptation and/or evolution. These findings highlight the importance of studying both sexes in epidemiological protocols and dietary interventions.


Asunto(s)
Alimentación Animal , Metilación de ADN , Grasas de la Dieta/metabolismo , Regulación de la Expresión Génica , Impresión Genómica , Placenta/metabolismo , Alelos , Animales , Epigénesis Genética , Femenino , Dosificación de Gen , Masculino , Ratones , Embarazo , Preñez , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...