Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Blood ; 127(1): 122-31, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26531164

RESUMO

T-cell-directed killing of tumor cells using bispecific antibodies is a promising approach for the treatment of hematologic malignancies. Here we describe our preclinical work with a dual-affinity retargeting (DART) molecule generated from antibodies to CD3 and CD123, designed to redirect T cells against acute myeloid leukemia blasts. The CD3×CD123 DART (also referred to as MGD006/S80880) consists of 2 independent polypeptides, each composed of the VH of 1 antibody in tandem with the VL of the other antibody. The target antigen CD123 (interleukin 3RA) is highly and differentially expressed in acute myeloid leukemia (AML) blasts compared with normal hematopoietic stem and progenitor cells. In this study we demonstrate that the CD3×CD123 DART binds to both human CD3 and CD123 to mediate target-effector cell association, T-cell activation, proliferation, and receptor diversification. The CD3×CD123 DART also induces a dose-dependent killing of AML cell lines and primary AML blasts in vitro and in vivo. These results provide the basis for testing the CD3×CD123 DART in the treatment of patients with CD123(+) AML.


Assuntos
Anticorpos Biespecíficos/imunologia , Apoptose , Complexo CD3/imunologia , Subunidade alfa de Receptor de Interleucina-3/imunologia , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Linfócitos T/imunologia , Animais , Complexo CD3/metabolismo , Proliferação de Células , Citometria de Fluxo , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T/genética , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Técnicas Imunoenzimáticas , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Cancer ; 13: 50, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24602489

RESUMO

BACKGROUND: Drug resistance is a significant problem in the treatment of ovarian cancer and can be caused by multiple mechanisms. Inhibition of apoptosis by the inhibitor of apoptosis proteins (IAPs) represents one such mechanism, and can be overcome by a mitochondrial protein called second mitochondria-derived activator of caspases (SMAC). We have previously shown that the ligands of sigma-2 receptors effectively induce tumor cell death. Additionally, because sigma-2 receptors are preferentially expressed in tumor cells, their ligands provide an effective mechanism for selective anti-cancer therapy. METHODS: In the current work, we have improved upon the previously described sigma-2 ligand SW43 by conjugating it to a pro-apoptotic small molecule SMAC mimetic SW IV-52, thus generating the novel cancer therapeutic SW IV-134. The new cancer drug was tested for receptor selectivity and tumor cell killing activity in vitro and in vivo. RESULTS: We have shown that SW IV-134 retained adequate sigma-2 receptor binding affinity in the context of the conjugate and potently induced cell death in ovarian cancer cells. The cell death induced by SW IV-134 was significantly greater than that observed with either SW43 or SW IV-52 alone and in combination. Furthermore, the intraperitoneal administration of SW IV-134 significantly reduced tumor burden and improved overall survival in a mouse xenograft model of ovarian cancer without causing significant adverse effects to normal tissues. Mechanistically, SW IV-134 induced degradation of cIAP-1 and cIAP-2 leading to NF-қB activation and TNFα-dependent cell death. CONCLUSIONS: Our findings suggest that coupling sigma-2 ligands to SMAC peptidomimetics enhances their effectiveness while maintaining the cancer selectivity. This encouraging proof-of-principle preclinical study supports further development of tumor-targeted small peptide mimetics via ligands to the sigma-2 receptor for future clinical applications.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Compostos Azabicíclicos/química , Compostos Azabicíclicos/farmacologia , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Receptores sigma/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Feminino , Xenoenxertos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Ligantes , Camundongos , Camundongos SCID , Proteínas Mitocondriais/química , Terapia de Alvo Molecular/métodos , Neoplasias Ovarianas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Blood ; 120(19): 4093-103, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22972985

RESUMO

The clinical goal of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is to minimize GVHD while maintaining GvL. Here, we show that interferon γ receptor-deficient (IFNγR(-/-)) allogeneic Tconv, which possess normal alloreactivity and cytotoxicity, induce significantly less GVHD than wild-type (WT) Tconv. This effect is mediated by altered trafficking of IFNγR(-/-) Tconv to GVHD target organs, especially the gastrointestinal (GI) tract. We show that the chemokine receptor CXCR3 is induced via IFNγR-mediated signaling and partially contributes to the trafficking of WT Tconv to GVHD target organs. Indeed, CXCR3(-/-) Tconv recapitulate the reduced GVHD potential of IFNγR(-/-) Tconv in a minor-mismatched GVHD model. Most importantly, IFNγR(-/-) (and CXCR3(-/-)) Tconv mediate a robust and beneficial GvL effect. In addition, we show that IFNγR(-/-) regulatory T cells (Tregs) are fully suppressive in vitro although defective in suppressor function in vivo and that WT Tregs suppress GVHD in vivo only when allogeneic Tconv produce interferon γ (IFNγ), suggesting that the IFNγR signaling pathway is the major mechanism for both Tregs and Tconv to migrate to GVHD target organs. Finally, pharmacologic inhibition of IFNγR signaling with inhibitors of JAK1/JAK2, which are mediators of IFNγR signaling, results in the decreased expression of CXCR3 and reduced GVHD and improved survival after allo-HSCT and this effect is mediated by altered trafficking of Tconv to GVHD target organs.


Assuntos
Movimento Celular/imunologia , Doença Enxerto-Hospedeiro/imunologia , Receptores de Interferon/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Cultivadas , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/mortalidade , Transplante de Células-Tronco Hematopoéticas , Humanos , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Nitrilas , Pirazóis/farmacologia , Pirimidinas , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Receptores CXCR3/metabolismo , Receptores de Interferon/genética , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Homólogo/efeitos adversos , Transplante Homólogo/imunologia , Receptor de Interferon gama
4.
BMC Cancer ; 14: 35, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24447304

RESUMO

BACKGROUND: The targeted delivery of cancer therapeutics represents an ongoing challenge in the field of drug development. TRAIL is a promising cancer drug but its activity profile could benefit from a cancer-selective delivery mechanism, which would reduce potential side effects and increase treatment efficiencies. We recently developed the novel TRAIL-based drug platform TR3, a genetically fused trimer with the capacity for further molecular modifications such as the addition of tumor-directed targeting moieties. MUC16 (CA125) is a well characterized biomarker in several human malignancies including ovarian, pancreatic and breast cancer. Mesothelin is known to interact with MUC16 with high affinity. In order to deliver TR3 selectively to MUC16-expressing cancers, we investigated the possibility of targeted TR3 delivery employing the high affinity mesothelin/MUC16 ligand/receptor interaction. METHODS: Using genetic engineering, we designed the novel cancer drug Meso-TR3, a fusion protein between native mesothelin and TR3. The recombinant proteins were produced with mammalian HEK293T cells. Meso-TR3 was characterized for binding selectivity and killing efficacy against MUC16-positive cancer cells and controls that lack MUC16 expression. Drug efficacy experiments were performed in vitro and in vivo employing an intraperitoneal xenograft mouse model of ovarian cancer. RESULTS: Similar to soluble mesothelin itself, the strong MUC16 binding property was retained in the Meso-TR3 fusion protein. The high affinity ligand/receptor interaction was associated with a selective accumulation of the cancer drug on MUC16-expressing cancer targets and directly correlated with increased killing activity in vitro and in a xenograft mouse model of ovarian cancer. The relevance of the mesothelin/MUC16 interaction for attaching Meso-TR3 to the cancer cells was verified by competitive blocking experiments using soluble mesothelin. Mechanistic studies using soluble DR5-Fc and caspase blocking assays confirmed engagement of the extrinsic death receptor pathway. Compared to non-targeted TR3, Meso-TR3 displayed a much reduced killing potency on cells that lack MUC16. CONCLUSIONS: Soluble Meso-TR3 targets the cancer biomarker MUC16 in vitro and in vivo. Following attachment to the tumor via surface bound MUC16, Meso-TR3 acquires full activation with superior killing profiles compared to non-targeted TR3, while its bioactivity is substantially reduced on cells that lack the tumor marker. This prodrug phenomenon represents a highly desirable property because it has the potential to enhance cancer killing with fewer side-effects than non-targeted TRAIL-based therapeutics. Thus, further exploration of this novel fusion protein is warranted as a possible therapeutic for patients with MUC16-positive malignancies.


Assuntos
Antineoplásicos/farmacologia , Antígeno Ca-125/metabolismo , Portadores de Fármacos , Proteínas Ligadas por GPI/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Antineoplásicos/metabolismo , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Ligantes , Mesotelina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Curr Biol ; 16(22): 2222-7, 2006 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-17113386

RESUMO

In eukaryotes, distinct regions of the genome are packaged as euchromatin (less condensed, more active) or heterochromatin (condensed, silenced). Studies in yeast, plants, and flies suggest that RNA interference (RNAi) is linked to heterochromatin formation and transcriptional silencing of transposable element (TE) sequences. We previously reported that insertion of a mobile hsp70-white reporter within 10 kb of a 1360 element on chromosome four of Drosophila melanogaster correlates with variegation (silencing). Here, we report small RNAs (approximately 23 nt) corresponding to 1360, indicating processing by the RNAi machinery. To directly test the ability of 1360 to silence a nearby gene in vivo, we introduced a P element construct carrying a single copy of 1360 upstream of the hsp70-white reporter into flies. This 1360 element contributes to HP1-dependent variegation at a pericentric insertion site, as demonstrated by a decrease in silencing after FLP-mediated removal of 1360. In euchromatin, 1360 is not sufficient to induce silencing, suggesting that proximity to pericentric heterochromatin and/or a high local TE density contributes to heterochromatin formation. Silencing of the 1360, hsp70-white reporter is sensitive to mutations in RNAi components. Our results implicate 1360 as a target for sequence-specific heterochromatic silencing through an RNAi-dependent mechanism.


Assuntos
Elementos de DNA Transponíveis/genética , Drosophila melanogaster/genética , Inativação Gênica , Heterocromatina/genética , Interferência de RNA , Animais , Northern Blotting , Primers do DNA , Proteínas de Drosophila , Genes Reporter/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas Nucleares , Proteínas de Ligação a RNA
6.
Mol Imaging ; 8(5): 245-53, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19796602

RESUMO

Small-animal tumor models are essential for developing translational therapeutic strategies in oncology research, with imaging having an increasingly important role. Magnetic resonance imaging (MRI) offers tumor localization, volumetric measurement, and the potential for advanced physiologic imaging but is less well suited to high-throughput studies and has limited capacity to assess early tumor growth. Bioluminescence imaging (BLI) identifies tumors early, monitors tumor growth, and efficiently measures response to therapeutic intervention. Generally, BLI signals have been found to correlate well with magnetic resonance measurements of tumor volume. However, in our studies of small-animal models of malignant brain tumors, we have observed specific instances in which BLI data do not correlate with corresponding MRIs. These observations led us to hypothesize that use of BLI and MRI together, rather than in isolation, would allow more effective and efficient measures of tumor growth in preclinical studies. Herein we describe combining BLI and MRI studies to characterize tumor growth in a mouse model of glioblastoma. The results led us to suggest a cost-effective, multimodality strategy for selecting cohorts of animals with similar tumor growth patterns that improves the accuracy of longitudinal in vivo measurements of tumor growth and treatment response in preclinical therapeutic studies.


Assuntos
Neoplasias Encefálicas/patologia , Diagnóstico por Imagem/métodos , Medições Luminescentes/métodos , Imageamento por Ressonância Magnética/métodos , Animais , Linhagem Celular Tumoral , Feminino , Glioblastoma/patologia , Camundongos , Camundongos Endogâmicos BALB C
7.
Community Pract ; 82(4): 20-3, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19397079

RESUMO

Health visitors identified fathers as a marginalised, difficult-to-reach group when aiming for universal implementation of a public health programme to prevent non-accidental head injury (NAHI) in babies. Follow-on research with 30 fathers from disadvantaged backgrounds, including some in prison, explored barriers and facilitating factors to preventing NAHI through focus groups and interviews. Fathers expressed both responsibility and helplessness in managing a baby, but many felt excluded from gaining skills and knowledge by healthcare staff. Barriers to implementing a prevention programme included a lack of knowledge about head injuries in babies and poor understanding of prevention as a reduction of risk factors. Fear of blame for a head injury could lead to injury concealment, indicating a taboo subject. Facilitators for fathers to learn about preventing head injuries in babies included concerns of their masculinity being at odds with the frailty of the newborn, recognition of fathers' needs to be valued in their own right, and a need for individualised plans for gaining information and increasing confidence. The modifiable barriers to prevention, from the fathers' perspectives, add to the theoretical and applied evidence base for the prevention of NAHI.


Assuntos
Pai , Educação em Saúde , Avaliação das Necessidades , Carência Psicossocial , Síndrome do Bebê Sacudido/prevenção & controle , Enfermagem em Saúde Comunitária , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Lactente , Masculino , Prisioneiros , País de Gales
8.
Mol Cell Biol ; 24(18): 8210-20, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15340080

RESUMO

The heterochromatic domains of Drosophila melanogaster (pericentric heterochromatin, telomeres, and the fourth chromosome) are characterized by histone hypoacetylation, high levels of histone H3 methylated on lysine 9 (H3-mK9), and association with heterochromatin protein 1 (HP1). While the specific interaction of HP1 with both H3-mK9 and histone methyltransferases suggests a mechanism for the maintenance of heterochromatin, it leaves open the question of how heterochromatin formation is targeted to specific domains. Expression characteristics of reporter transgenes inserted at different sites in the fourth chromosome define a minimum of three euchromatic and three heterochromatic domains, interspersed. Here we searched for cis-acting DNA sequence determinants that specify heterochromatic domains. Genetic screens for a switch in phenotype demonstrate that local deletions or duplications of 5 to 80 kb of DNA flanking a transposon reporter can lead to the loss or acquisition of variegation, pointing to short-range cis-acting determinants for silencing. This silencing is dependent on HP1. A switch in transgene expression correlates with a switch in chromatin structure, judged by nuclease accessibility. Mapping data implicate the 1360 transposon as a target for heterochromatin formation. We propose that heterochromatin formation is initiated at dispersed repetitive elements along the fourth chromosome and spreads for approximately 10 kb or until encountering competition from a euchromatic determinant.


Assuntos
Drosophila melanogaster/genética , Heterocromatina/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Mapeamento Cromossômico , Cromossomos/genética , DNA/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Anormalidades do Olho/genética , Feminino , Inativação Gênica , Genes de Insetos , Genes Reporter , Teste de Complementação Genética , Heterocromatina/metabolismo , Masculino , Mutação , Fenótipo , Deleção de Sequência , Sequências de Repetição em Tandem
9.
Am J Obstet Gynecol ; 194(4): 1137-42; discussion 1142-4, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16580313

RESUMO

OBJECTIVE: The purpose of this study was to test the hypothesis that placental samples that are obtained by needle aspiration ex vivo are useful for the determination of villus gene expression. STUDY DESIGN: Placental biopsy was performed with a spinal needle after uncomplicated deliveries. Villi were inspected microscopically, and RNA was extracted and analyzed with capillary electrophoresis. Gene expression was determined with quantitative polymerase chain reaction. RESULTS: We obtained more placental villous fragments per aspiration using a 20-gauge needle (5.2 +/- 1.8 fragments) than with a 22-gauge needle (3.3 +/- 1.6 fragments; P < .01). RNA quality was adequate, based on the 28S and 18S recombinant RNA bands, with a mean 260/280 ratio of 1.88. The amount of extracted RNA correlated with the number of villous fragments per aspirate. Importantly, the expression of NDRG1 and hPL, both markedly altered in hypoxia, was consistent between villi that were obtained by either needle or standard biopsy. CONCLUSION: Placental samples that are obtained by ex vivo needle aspiration are useful for the extraction of RNA and for the determination of villous gene expression.


Assuntos
Biópsia por Agulha , Placenta/química , RNA/análise , Proteínas de Ciclo Celular , Feminino , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lactogênio Placentário/genética , Reação em Cadeia da Polimerase , Proteínas/genética , RNA/isolamento & purificação
10.
Clin Cancer Res ; 22(3): 596-608, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26423796

RESUMO

PURPOSE: Anti-CD20 monoclonal antibodies (mAb) are an important immunotherapy for B-cell lymphoma, and provide evidence that the immune system may be harnessed as an effective lymphoma treatment approach. ALT-803 is a superagonist IL-15 mutant and IL-15Rα-Fc fusion complex that activates the IL-15 receptor constitutively expressed on natural killer (NK) cells. We hypothesized that ALT-803 would enhance anti-CD20 mAb-directed NK-cell responses and antibody-dependent cellular cytotoxicity (ADCC). EXPERIMENTAL DESIGN: We tested this hypothesis by adding ALT-803 immunostimulation to anti-CD20 mAb triggering of NK cells in vitro and in vivo. Cell lines and primary human lymphoma cells were utilized as targets for primary human NK cells. Two complementary in vivo mouse models were used, which included human NK-cell xenografts in NOD/SCID-γc (-/-) mice. RESULTS: We demonstrate that short-term ALT-803 stimulation significantly increased degranulation, IFNγ production, and ADCC by human NK cells against B-cell lymphoma cell lines or primary follicular lymphoma cells. ALT-803 augmented cytotoxicity and the expression of granzyme B and perforin, providing one potential mechanism for this enhanced functionality. Moreover, in two distinct in vivo B-cell lymphoma models, the addition of ALT-803 to anti-CD20 mAb therapy resulted in significantly reduced tumor cell burden and increased survival. Long-term ALT-803 stimulation of human NK cells induced proliferation and NK-cell subset changes with preserved ADCC. CONCLUSIONS: ALT-803 represents a novel immunostimulatory drug that enhances NK-cell antilymphoma responses in vitro and in vivo, thereby supporting the clinical investigation of ALT-803 plus anti-CD20 mAbs in patients with indolent B-cell lymphoma.


Assuntos
Antineoplásicos/farmacologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Proteínas/farmacologia , Receptores de IgG/metabolismo , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Linhagem Celular Tumoral , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Granzimas/genética , Granzimas/metabolismo , Humanos , Interferon gama/biossíntese , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/patologia , Camundongos , Camundongos Knockout , Perforina/genética , Perforina/metabolismo , Proteínas Recombinantes de Fusão , Rituximab/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Discov ; 4(7): 828-39, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24740997

RESUMO

UNLABELLED: Recently identified isocitrate dehydrogenase (IDH) mutations lead to the production of 2-hydroxyglutarate (2HG), an oncometabolite aberrantly elevated in selected cancers. We developed a facile and inexpensive fluorimetric microplate assay for the quantitation of 2HG and performed an unbiased small-molecule screen in live cells to identify compounds capable of perturbing 2HG production. Zaprinast, a phosphodiesterase 5 inhibitor, was identified as an efficacious modulator of 2HG production and confirmed to lower 2HG levels in vivo. The mechanism of action was not due to cGMP stabilization, but rather, profiling of metabolites upstream of mutant IDH1 pointed to targeted inhibition of the enzyme glutaminase (GLS). Zaprinast treatment reversed histone hypermethylation and soft-agar growth of IDH1-mutant cells, and treatment of glutamine-addicted pancreatic cancer cells reduced growth and sensitized cells to oxidative damage. Thus, Zaprinast is efficacious against glutamine metabolism and further establishes the therapeutic linkages between GLS and 2HG-mediated oncogenesis. SIGNIFICANCE: Gain-of-function IDH mutations are common events in glioma, acute myelogenous leukemia, and other cancer types, which lead to the accumulation of the oncometabolite 2HG. We show that the drug Zaprinast is capable of reducing cellular 2HG levels by inhibiting the upstream enzyme GLS, thus identifying a new strategy to target 2HG production in selected IDH-mutant cancers.


Assuntos
Inibidores Enzimáticos/farmacologia , Fibrossarcoma/tratamento farmacológico , Isocitrato Desidrogenase/metabolismo , Purinonas/farmacologia , Animais , Linhagem Celular Tumoral , Fibrossarcoma/enzimologia , Glutaminase , Glutaratos/metabolismo , Células HEK293 , Ensaios de Triagem em Larga Escala , Histonas , Humanos , Metilação , Camundongos , Camundongos Nus , Neoplasias Experimentais
12.
PLoS One ; 9(10): e109799, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25289677

RESUMO

We have recently reported that interferon gamma receptor deficient (IFNγR-/-) allogeneic donor T cells result in significantly less graft-versus-host disease (GvHD) than wild-type (WT) T cells, while maintaining an anti-leukemia or graft-versus-leukemia (GvL) effect after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We demonstrated that IFNγR signaling regulates alloreactive T cell trafficking to GvHD target organs through expression of the chemokine receptor CXCR3 in alloreactive T cells. Since IFNγR signaling is mediated via JAK1/JAK2, we tested the effect of JAK1/JAK2 inhibition on GvHD. While we demonstrated that pharmacologic blockade of JAK1/JAK2 in WT T cells using the JAK1/JAK2 inhibitor, INCB018424 (Ruxolitinib), resulted in a similar effect to IFNγR-/- T cells both in vitro (reduction of CXCR3 expression in T cells) and in vivo (mitigation of GvHD after allo-HSCT), it remains to be determined if in vivo administration of INCB018424 will result in preservation of GvL while reducing GvHD. Here, we report that INCB018424 reduces GvHD and preserves the beneficial GvL effect in two different murine MHC-mismatched allo-HSCT models and using two different murine leukemia models (lymphoid leukemia and myeloid leukemia). In addition, prolonged administration of INCB018424 further improves survival after allo-HSCT and is superior to other JAK1/JAK2 inhibitors, such as TG101348 or AZD1480. These data suggest that pharmacologic inhibition of JAK1/JAK2 might be a promising therapeutic approach to achieve the beneficial anti-leukemia effect and overcome HLA-barriers in allo-HSCT. It might also be exploited in other diseases besides GvHD, such as organ transplant rejection, chronic inflammatory diseases and autoimmune diseases.


Assuntos
Regulação Leucêmica da Expressão Gênica , Doença Enxerto-Hospedeiro/prevenção & controle , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Leucemia Linfoide/tratamento farmacológico , Leucemia Mieloide/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Animais , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/enzimologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Efeito Enxerto vs Leucemia/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Janus Quinase 1/genética , Janus Quinase 1/imunologia , Janus Quinase 2/genética , Janus Quinase 2/imunologia , Leucemia Linfoide/enzimologia , Leucemia Linfoide/imunologia , Leucemia Linfoide/patologia , Leucemia Mieloide/enzimologia , Leucemia Mieloide/imunologia , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Nitrilas , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirrolidinas/farmacologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Transdução de Sinais , Sulfonamidas/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/enzimologia , Linfócitos T/imunologia , Linfócitos T/patologia , Transplante Homólogo , Irradiação Corporal Total , Receptor de Interferon gama
13.
Cancer Discov ; 3(2): 212-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23269702

RESUMO

UNLABELLED: Inhibitor of apoptosis (IAP) proteins play a central role in many types of cancer, and IAP antagonists are in development as anticancer agents. IAP antagonists cause apoptosis in many cells, but they also activate alternative NF-κB signaling through NF-κB-inducing kinase (NIK), which regulates osteoclasts. In bone metastasis, a positive feedback loop between tumors and osteoclasts promotes tumor growth and osteolysis. We therefore tested the effect of IAP antagonists on the bone microenvironment for metastasis. In both drug-sensitive and drug-resistant tumors, growth in bone was favored, as compared with other sites during IAP antagonist treatment. These drugs also caused osteoporosis and increased osteoclastogenesis, mediated by NIK, and enhanced tumor-associated osteolysis. Cotreatment with zoledronic acid, a potent osteoclast inhibitor, reduced IAP antagonist-enhanced tumor growth in bone and osteolysis. Thus, IAP antagonist-based cancer treatment may be compromised by osteoporosis and enhanced skeletal metastasis, which may be prevented by antiresorptive agents. SIGNIFICANCE: Although IAP antagonists are a class of anticancer agents with proven efficacy in multiple cancers, we show that these agents can paradoxically increase tumor growth and metastasis in the bone by stabilizing NIK and activating the alternative NF-κB pathway in osteoclasts. Future clinical trials of IAP antagonist-based therapy may require detailed examination of this potential for enhanced bone metastasis and osteoporosis, as well as possible combination with antiresorptive agents.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Neoplasias Ósseas/secundário , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Osteoclastos/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/química , Neoplasias Ósseas/metabolismo , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Difosfonatos/administração & dosagem , Difosfonatos/farmacologia , Humanos , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Proteínas Inibidoras de Apoptose/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Estrutura Molecular , Oligopeptídeos/administração & dosagem , Oligopeptídeos/farmacologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteoporose/induzido quimicamente , Osteoporose/prevenção & controle , Ensaios Antitumorais Modelo de Xenoenxerto , Ácido Zoledrônico
14.
Cancer Res ; 73(3): 1128-41, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23221383

RESUMO

Tumor-infiltrating immune cells can promote chemoresistance and metastatic spread in aggressive tumors. Consequently, the type and quality of immune responses present in the neoplastic stroma are highly predictive of patient outcome in several cancer types. In addition to host immune responses, intrinsic tumor cell activities that mimic stem cell properties have been linked to chemoresistance, metastatic dissemination, and the induction of immune suppression. Cancer stem cells are far from a static cell population; rather, their presence seems to be controlled by highly dynamic processes that are dependent on cues from the tumor stroma. However, the impact immune responses have on tumor stem cell differentiation or expansion is not well understood. In this study, we show that targeting tumor-infiltrating macrophages (TAM) and inflammatory monocytes by inhibiting either the myeloid cell receptors colony-stimulating factor-1 receptor (CSF1R) or chemokine (C-C motif) receptor 2 (CCR2) decreases the number of tumor-initiating cells (TIC) in pancreatic tumors. Targeting CCR2 or CSF1R improves chemotherapeutic efficacy, inhibits metastasis, and increases antitumor T-cell responses. Tumor-educated macrophages also directly enhanced the tumor-initiating capacity of pancreatic tumor cells by activating the transcription factor STAT3, thereby facilitating macrophage-mediated suppression of CD8(+) T lymphocytes. Together, our findings show how targeting TAMs can effectively overcome therapeutic resistance mediated by TICs.


Assuntos
Tolerância Imunológica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Receptores CCR2/antagonistas & inibidores , Animais , Comunicação Celular , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Macrófagos/fisiologia , Camundongos , Metástase Neoplásica , Neoplasias/imunologia , Neoplasias/patologia , Receptor de Fator Estimulador de Colônias de Macrófagos/fisiologia , Receptores CCR2/fisiologia , Fator de Transcrição STAT3/fisiologia , Linfócitos T Citotóxicos/imunologia , Gencitabina
15.
J Clin Invest ; 122(10): 3579-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996695

RESUMO

The adenosine diphosphate (ADP) receptor P2RY12 (purinergic receptor P2Y, G protein coupled, 12) plays a critical role in platelet aggregation, and P2RY12 inhibitors are used clinically to prevent cardiac and cerebral thrombotic events. Extracellular ADP has also been shown to increase osteoclast (OC) activity, but the role of P2RY12 in OC biology is unknown. Here, we examined the role of mouse P2RY12 in OC function. Mice lacking P2ry12 had decreased OC activity and were partially protected from age-associated bone loss. P2ry12-/- OCs exhibited intact differentiation markers, but diminished resorptive function. Extracellular ADP enhanced OC adhesion and resorptive activity of WT, but not P2ry12-/-, OCs. In platelets, ADP stimulation of P2RY12 resulted in GTPase Ras-related protein (RAP1) activation and subsequent αIIbß3 integrin activation. Likewise, we found that ADP stimulation induced RAP1 activation in WT and integrin ß3 gene knockout (Itgb3-/-) OCs, but its effects were substantially blunted in P2ry12-/- OCs. In vivo, P2ry12-/- mice were partially protected from pathologic bone loss associated with serum transfer arthritis, tumor growth in bone, and ovariectomy-induced osteoporosis: all conditions associated with increased extracellular ADP. Finally, mice treated with the clinical inhibitor of P2RY12, clopidogrel, were protected from pathologic osteolysis. These results demonstrate that P2RY12 is the primary ADP receptor in OCs and suggest that P2RY12 inhibition is a potential therapeutic target for pathologic bone loss.


Assuntos
Difosfato de Adenosina/fisiologia , Remodelação Óssea/fisiologia , Osteoclastos/fisiologia , Osteoporose/fisiopatologia , Receptores Purinérgicos P2Y12/fisiologia , Animais , Artrite Experimental/complicações , Neoplasias Ósseas/complicações , Neoplasias Ósseas/secundário , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/fisiopatologia , Carcinoma/complicações , Carcinoma/secundário , Adesão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Clopidogrel , Ativação Enzimática/efeitos dos fármacos , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoporose/etiologia , Osteoporose/prevenção & controle , Ovariectomia , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y12/deficiência , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/genética , Organismos Livres de Patógenos Específicos , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico , Proteínas rap1 de Ligação ao GTP/efeitos dos fármacos
16.
Cancer Res ; 71(14): 4799-808, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21602433

RESUMO

Blockade of osteoclast (OC) activity efficiently decreases tumor burden as well as associated bone erosion in immune-compromised animals bearing human osteolytic cancers. In this study, we showed that modulation of antitumor T-cell responses alters tumor growth in bone, regardless of OC status, by using genetic and pharmacologic models. PLCγ2(-/-) mice, with dysfunctional OCs and impaired dendritic cell (DC)-mediated T-cell activation, had increased bone tumor burden despite protection from bone loss. In contrast, Lyn(-/-) mice, with more numerous OCs and a hyperactive myeloid population leading to increased T-cell responses, had reduced tumor growth in bone despite enhanced osteolysis. The unexpected tumor/bone phenotype observed in PLCγ2(-/-) and Lyn(-/-) mice was transplantable, suggesting the involvement of an immune component. Consistent with this hypothesis, T-cell activation diminished skeletal metastasis whereas T-cell depletion enhanced it, even in the presence of zoledronic acid, a potent antiresorptive agent. Importantly, injection of antigen-specific wild-type cytotoxic CD8(+) T cells in PLCγ2(-/-) mice or CD8(+) T-cell depletion in Lyn(-/-) mice normalized tumor growth in bone. Our findings show the important contribution of CD8(+) T cells in the regulation of bone metastases regardless of OC status, thus including T cells as critical regulators of tumor growth in bone.


Assuntos
Neoplasias Ósseas/imunologia , Reabsorção Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Osteoclastos/imunologia , Animais , Neoplasias Ósseas/enzimologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Reabsorção Óssea/enzimologia , Reabsorção Óssea/patologia , Processos de Crescimento Celular/imunologia , Feminino , Imunoterapia Adotiva , Ativação Linfocitária , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoclastos/enzimologia , Osteoclastos/patologia , Fosfolipase C gama/deficiência , Fosfolipase C gama/imunologia , Quinases da Família src/deficiência , Quinases da Família src/imunologia
17.
PLoS One ; 6(9): e24018, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21915277

RESUMO

Pivotal components of the IFN response to virus infection include the IFN receptors (IFNR), and the downstream factor signal transducer and activator of transcription 1 (Stat1). Mice deficient for Stat1 and IFNR (Stat1(-/-) and IFNαßγR(-/-) mice) lack responsiveness to IFN and exhibit high sensitivity to various pathogens. Here we examined herpes simplex virus type 1 (HSV-1) pathogenesis in Stat1(-/-) mice and in IFNαßγR(-/-) mice following corneal infection and bioluminescent imaging. Two divergent and paradoxical patterns of infection were observed. Mice with an N-terminal deletion in Stat1 (129Stat1(-/-) (N-term)) had transient infection of the liver and spleen, but succumbed to encephalitis by day 10 post-infection. In stark contrast, infection of IFNαßγR(-/-) mice was rapidly fatal, with associated viremia and fulminant infection of the liver and spleen, with infected infiltrating cells being primarily of the monocyte/macrophage lineage. To resolve the surprising difference between Stat1(-/-) and IFNαßγR(-/-) mice, we infected an additional Stat1(-/-) strain deleted in the DNA-binding domain (129Stat1(-/-) (DBD)). These 129Stat1(-/-) (DBD) mice recapitulated the lethal pattern of liver and spleen infection seen following infection of IFNαßγR(-/-) mice. This lethal pattern was also observed when 129Stat1(-/-) (N-term) mice were infected and treated with a Type I IFN-blocking antibody, and immune cells derived from 129Stat1(-/-) (N-term) mice were shown to be responsive to Type I IFN. These data therefore show significant differences in viral pathogenesis between two commonly-used Stat1(-/-) mouse strains. The data are consistent with the hypothesis that Stat1(-/-) (N-term) mice have residual Type I IFN receptor-dependent IFN responses. Complete loss of IFN signaling pathways allows viremia and rapid viral spread with a fatal infection of the liver. This study underscores the importance of careful comparisons between knockout mouse strains in viral pathogenesis, and may also be relevant to the causation of HSV hepatitis in humans, a rare but frequently fatal infection.


Assuntos
Herpes Simples/patologia , Medições Luminescentes/métodos , Receptores de Interferon/deficiência , Fator de Transcrição STAT1/deficiência , Animais , Herpes Simples/genética , Herpes Simples/metabolismo , Herpesvirus Humano 1/patogenicidade , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptores de Interferon/genética , Fator de Transcrição STAT1/genética , Baço/patologia , Baço/virologia
18.
Cancer Res ; 69(22): 8700-9, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19843867

RESUMO

To define the factors that modulate regulatory T (Treg) cells in the tumor setting, we cocultured various tumor cells with either purified Treg cells, or with unfractionated splenocytes. We found that Treg expansion occurred only with unfractionated splenocytes, suggesting that accessory cells and/or factors produced by them play an essential role in tumor-induced Treg expansion. We performed gene expression profiling on tumor-associated Treg cells to identify candidate signaling molecules and studied their effects on tumor-induced Treg expansion. We inadvertently discovered that interleukin (IL)-12 treatment blocked Treg expansion in an IL-12 receptor-dependent fashion. Additional studies showed that IL-12 acts by stimulating IFN-gamma mediated inhibition of Treg cell proliferation, which may partially account for the antitumor effects of IL-12. Furthermore, IL-12 treatment was found to decrease IL-2 production, which may lead to IFN-gamma-independent inhibition of Treg cells, as IL-2 is required for their survival and expansion. Mechanistic studies revealed that IFN-gamma signaling directly causes cell cycle arrest in Treg cells. This study shows that an IL-12-IFN-gamma axis can suppress tumor-induced Treg proliferation. This mechanism may counteract the ability of Treg cells to promote tumor growth in vivo.


Assuntos
Perfilação da Expressão Gênica , Interferon gama/imunologia , Interleucina-12/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Citometria de Fluxo , Expressão Gênica , Interferon gama/metabolismo , Interleucina-12/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Linfócitos T Reguladores/metabolismo
19.
Immunity ; 27(4): 635-46, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17919943

RESUMO

Granzyme B is important for the ability of NK cells and CD8(+) T cells to kill their targets. However, we showed here that granzyme B-deficient mice clear both allogeneic and syngeneic tumor cell lines more efficiently than do wild-type (WT) mice. To determine whether regulatory T (Treg) cells utilize granzyme B to suppress immune responses against these tumors, we examined the expression and function of granzyme B in Treg cells. Granzyme B was not expressed in naive Treg cells but was highly expressed in 5%-30% of CD4(+)Foxp3(+) Treg cells in the tumor environment. Adoptive transfer of WT Treg cells, but not granzyme B- or perforin-deficient Treg cells, into granzyme B-deficient mice partially restored susceptibility to tumor growth; Treg cells derived from the tumor environment could induce NK and CD8(+) T cell death in a granzyme B- and perforin-dependent fashion. Granzyme B and perforin are therefore relevant for Treg cell-mediated suppression of tumor clearance in vivo.


Assuntos
Citotoxicidade Imunológica , Granzimas/imunologia , Subpopulações de Linfócitos/imunologia , Neoplasias Experimentais/imunologia , Perforina/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Granzimas/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Subpopulações de Linfócitos/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Congênicos , Modelos Imunológicos , Perforina/metabolismo , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA