Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Br J Cancer ; 113(11): 1541-7, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26554652

RESUMEN

BACKGROUND: PI3K-AKT-mTOR inhibitors (PAMi) are promising anticancer treatments. Hyperglycaemia is a mechanism-based toxicity of these agents and is becoming increasingly important with their use in larger numbers of patients. METHODS: Retrospective case-control study comparing incidence and severity of hyperglycaemia (all grades) between a case group of 387 patients treated on 18 phase I clinical trials with PAMi (78 patients with PI3Ki, 138 with mTORi, 144 with AKTi and 27 with PI3K/mTORi) and a control group of 109 patients treated on 10 phase I clinical trials with agents not directly targeting the PAM pathway. Diabetic patients were excluded in both groups. RESULTS: The incidence of hyperglycaemia was not significantly different between cases and controls (86.6% vs 80.7%, respectively, P=0.129). However, high grade (grade 3-4) hyperglycaemia was more frequent in the PAMi group than in controls (6.7% vs 0%, respectively, P=0.005). The incidence of grade 3-4 hyperglycaemia was greater with AKT and multikinase inhibitors compared with other PAMi (P<0.001). All patients with high-grade hyperglycaemia received antihyperglycemic treatment and none developed severe metabolic complications (diabetic ketoacidosis or hyperosmolar hyperglycemic nonketotic state). High-grade hyperglycaemia was the cause of permanent PAMi discontinuation in nine patients. CONCLUSIONS: PI3K-AKT-mTOR inhibitors are associated with small (6.7%) but statistically significant increased risk of high-grade hyperglycaemia compared with non-PAM targeting agents. However, PAMi-induced hyperglycaemia was not found to be associated with severe metabolic complications in this non-diabetic population of patients with advanced cancers.


Asunto(s)
Antineoplásicos/efectos adversos , Hiperglucemia/epidemiología , Hiperglucemia/etiología , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Glucemia/efectos de los fármacos , Estudios de Casos y Controles , Ensayos Clínicos Fase I como Asunto , Femenino , Humanos , Hiperglucemia/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Masculino , Persona de Mediana Edad , Neoplasias/patología , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Adulto Joven
2.
Ann Oncol ; 24(6): 1491-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23406736

RESUMEN

BACKGROUND: Bone marrow-derived endothelial progenitor cells (EPCs) are critical for metastatic progression. This study explores the effect of tetrathiomolybdate (TM), an anti-angiogenic copper chelator, on EPCs in patients at high risk for breast cancer recurrence. PATIENTS AND METHODS: This phase 2 study enrolled breast cancer patients with stage 3 and stage 4 without evidence of disease (NED), and stage 2 if triple-negative. TM 100 mg orally was administered to maintain ceruloplasmin <17 mg/dl for 2 years or until relapse. The primary end point was change in EPCs. RESULTS: Forty patients (28 stage 2/3, 12 stage 4 NED) were enrolled. Seventy-five percent patients achieved the copper depletion target by 1 month. Ninety-one percent of triple-negative patients copper-depleted compared with 41% luminal subtypes. In copper-depleted patients only, there was a significant reduction in EPCs/ml by 27 (P = 0.04). Six patients relapsed while on study, of which only one patient had EPCs maintained below baseline. The 10-month relapse-free survival was 85.0% (95% CI 74.6%-96.8%). Only grade 3/4 toxicity was hematologic: neutropenia (3.1% of cycles), febrile neutropenia (0.2%), and anemia (0.2%). CONCLUSIONS: TM is safe and appears to maintain EPCs below baseline in copper-depleted patients. TM may promote tumor dormancy and ultimately prevent relapse.


Asunto(s)
Neoplasias de la Mama/sangre , Cobre/sangre , Células Endoteliales/metabolismo , Molibdeno/uso terapéutico , Recurrencia Local de Neoplasia/prevención & control , Células Madre/metabolismo , Adulto , Anciano , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Quelantes/uso terapéutico , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Persona de Mediana Edad , Molibdeno/farmacología , Recurrencia Local de Neoplasia/sangre , Factores de Riesgo , Células Madre/efectos de los fármacos
3.
Nat Med ; 7(11): 1194-201, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11689883

RESUMEN

The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1+/-Id3-/- host, which were associated with VEGF-receptor-1-positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.


Asunto(s)
Células Madre Hematopoyéticas/patología , Proteínas de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neovascularización Patológica , Proteínas Represoras , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/patología , Trasplante de Células Madre Hematopoyéticas , Proteína 1 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutación , Neovascularización Patológica/genética , Pruebas de Neutralización , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular
4.
J Exp Med ; 170(6): 2105-18, 1989 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-2584937

RESUMEN

A pore-forming protein (PFP; perforin) and various serine esterases (SE) have been identified in the cytoplasmic granules of CTL and NK cells. Perforin and several SE have recently been cloned. Northern blotting analysis was performed here using cDNA probes specific for human perforin and two SE (SE 1/HS and SE 2/GB) to monitor the levels of specific mRNAs in mitogen-stimulated primary human T cells. These mRNAs were rapidly induced by IL-2 with optimal responses at 300 U/ml. After IL-2 treatment, mRNAs for perforin, SE 1, and SE 2 peaked at 12-24 h and decreased after 48 h. The three mRNAs were also induced in T cells treated with a combination of PMA plus lectin, OKT3 mAb, or plastic-adherent accessory cells. However, the induction induced by PMA/mitogen followed a slower kinetics, peaking at 48 h. In general, we found that SE 1 mRNA was more readily induced by IL-2, while SE 2 responded better to PMA/mitogen. Similar patterns of mRNA expression were observed for both unprimed T cells and PHA-primed T blasts. After stimulation with IL-2 and PMA/mitogen, the T8+ subset was shown to be the main producer of perforin, SE 1, and SE 2. Low levels of all three mRNAs, however, were also detected in the T4+ subset. The induction of all three mRNAs by either IL-2 or PMA/mitogen was partially blocked by the immunosuppressive drug cyclosporin A (CsA), but not by the biologically inactive analogue cyclosporin H. Together, these results point to some similarities and differences with upregulation of granule mediator mRNAs relative to lymphokine mRNAs. Both sets of genes require two signals for their induction by mitogens. In contrast to lymphokines, there is a strong response of granule mRNAs to IL-2, and the induction of these transcripts is only partially blocked by CsA.


Asunto(s)
Ciclosporinas/farmacología , Esterasas/genética , Activación de Linfocitos , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Linfocitos T/metabolismo , ADN/aislamiento & purificación , Expresión Génica , Humanos , Interleucina-2/farmacología , Cinética , Perforina , Proteínas Citotóxicas Formadoras de Poros , ARN Mensajero/análisis , Acetato de Tetradecanoilforbol/farmacología
5.
J Exp Med ; 183(5): 2385-90, 1996 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8642350

RESUMEN

Herpesviral DNA fragments isolated from AIDS-associated Kaposi's sarcoma (KS) tissue (KSHV-DNA) share homology with two lymphotropic oncogenic gamma-herpesviruses, Epstein-Barr virus and Herpesvirus saimiri, and are present in the lesions of more than 95% of HIV and non-HIV-associated forms of KS, AIDS-related body cavity-based lymphomas, and AIDS-related multicentric Castleman's disease. Here we show that BC-1, a KSHV-DNA-positive, body cavity-based lymphoma cell line, produces infective herpesviral particles carrying a linear 270-kb genome that specifically transmits KSHV-DNA to CD19+ B cells. Transmission of KSHV-DNA is dependent upon a biologically active, replicating virus, since it is blocked by UV irradiation and foscarnet, an inhibitor of viral DNA-polymerase. This study represents the first isolation and transmission of the human herpesvirus-8/KS-associated herpesvirus.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/complicaciones , Linfocitos B/virología , Herpesviridae/clasificación , Herpesviridae/fisiología , Sarcoma de Kaposi/virología , Southern Blotting , Línea Celular , Sondas de ADN , ADN Viral/análisis , Sangre Fetal , Genoma Viral , Herpesviridae/aislamiento & purificación , Herpesvirus Saimiriino 2/clasificación , Herpesvirus Humano 4/clasificación , Humanos , Reacción en Cadena de la Polimerasa , Sarcoma de Kaposi/etiología
6.
J Exp Med ; 186(8): 1247-56, 1997 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-9334364

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in the initiation of antitumor immune responses. In this study, we show that genetic modifications of a murine epidermis-derived DC line and primary bone marrow-derived DCs to express a model antigen beta-galactosidase (betagal) can be achieved through the use of a replication-deficient, recombinant adenovirus vector, and that the modified DCs are capable of eliciting antigen-specific, MHC-restricted CTL responses. Importantly, using a murine metastatic lung tumor model with syngeneic colon carcinoma cells expressing betagal, we show that immunization of mice with the genetically modified DC line or bone marrow DCs confers potent protection against a lethal tumor challenge, as well as suppression of preestablished tumors, resulting in a significant survival advantage. We conclude that genetic modification of DCs to express antigens that are also expressed in tumors can lead to antigen-specific, antitumor killer cells, with a concomitant resistance to tumor challenge and a decrease in the size of existing tumors.


Asunto(s)
Adenocarcinoma/inmunología , Adenoviridae/genética , Neoplasias del Colon/inmunología , ADN Complementario/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Inmunoterapia Adoptiva/métodos , beta-Galactosidasa/inmunología , Adenocarcinoma/prevención & control , Adenocarcinoma/terapia , Adenoviridae/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología , Trasplante de Médula Ósea , Línea Celular , Neoplasias del Colon/prevención & control , Neoplasias del Colon/terapia , Células Dendríticas/virología , Técnicas de Transferencia de Gen , Vectores Genéticos/inmunología , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Activación de Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Inmunológicos , Trasplante de Neoplasias , Linfocitos T Citotóxicos/inmunología
7.
J Exp Med ; 188(3): 539-48, 1998 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-9687531

RESUMEN

Although thrombopoietin has been shown to promote megakaryocyte (MK) proliferation and maturation, the exact mechanism and site of platelet formation are not well defined. Studies have shown that MKs may transmigrate through bone marrow endothelial cells (BMEC), and release platelets within the sinusoidal space or lung capillaries. In search for chemotactic factor(s) that may mediate transmigration of MKs, we have discovered that mature polyploid MKs express the G protein-coupled chemokine receptor CXCR4 (Fusin, LESTR). Therefore, we explored the possibility that stromal cell-derived factor 1 (SDF-1), the ligand for CXCR4, may also induce transendothelial migration of mature MKs. SDF-1, but not other CXC or CC chemokines, was able to mediate MK migration (ED50 = 125 pmol/liter). The MK chemotaxis induced by SDF-1 was inhibited by the CXCR4-specific mAb (12G5) and by pertussis toxin, demonstrating that signaling via the G protein-coupled receptor CXCR4 was necessary for migration. SDF-1 also induced MKs to migrate through confluent monolayers of BMEC by increasing the affinity of MKs for BMEC. Activation of BMEC with interleukin 1beta resulted in a threefold increase in the migration of MKs in response to SDF-1. Neutralizing mAb to the endothelial-specific adhesion molecule E-selectin blocked the migration of MKs by 50%, suggesting that cellular interaction of MKs with BMEC is critical for the migration of MKs. Light microscopy and ploidy determination of transmigrated MKs demonstrated predominance of polyploid MKs. Virtually all platelets generated in the lower chamber also expressed CXCR4. Platelets formed in the lower chamber were functional and expressed P-selectin (CD62P) in response to thrombin stimulation. Electron microscopy of the cells that transmigrated through the BMEC monolayers in response to SDF-1 demonstrated the presence of intact polyploid MKs as well as MKs in the process of platelet formation. These results suggest that SDF-1 is a potent chemotactic factor for mature MKs. Expression of CXCR4 may be the critical cellular signal for transmigration of MKs and platelet formation.


Asunto(s)
Plaquetas/fisiología , Médula Ósea/fisiología , Quimiocinas CXC/fisiología , Quimiotaxis/fisiología , Endotelio Vascular/fisiología , Megacariocitos/fisiología , Línea Celular , Quimiocina CXCL12 , Humanos , Megacariocitos/efectos de los fármacos , Poliploidía , Receptores CXCR4/biosíntesis
8.
J Exp Med ; 193(9): 1005-14, 2001 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-11342585

RESUMEN

Tyrosine kinase receptors for angiogenic factors vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) are expressed not only by endothelial cells but also by subsets of hematopoietic stem cells (HSCs). To further define their role in the regulation of postnatal hematopoiesis and vasculogenesis, VEGF and Ang-1 plasma levels were elevated by injecting recombinant protein or adenoviral vectors expressing soluble VEGF(165), matrix-bound VEGF(189), or Ang-1 into mice. VEGF(165), but not VEGF(189), induced a rapid mobilization of HSCs and VEGF receptor (VEGFR)2(+) circulating endothelial precursor cells (CEPs). In contrast, Ang-1 induced delayed mobilization of CEPs and HSCs. Combined sustained elevation of Ang-1 and VEGF(165) was associated with an induction of hematopoiesis and increased marrow cellularity followed by proliferation of capillaries and expansion of sinusoidal space. Concomitant to this vascular remodeling, there was a transient depletion of hematopoietic activity in the marrow, which was compensated by an increase in mobilization and recruitment of HSCs and CEPs to the spleen resulting in splenomegaly. Neutralizing monoclonal antibody to VEGFR2 completely inhibited VEGF(165), but not Ang-1-induced mobilization and splenomegaly. These data suggest that temporal and regional activation of VEGF/VEGFR2 and Ang-1/Tie-2 signaling pathways are critical for mobilization and recruitment of HSCs and CEPs and may play a role in the physiology of postnatal angiogenesis and hematopoiesis.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Linfocinas/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Adenoviridae , Angiopoyetina 1 , Animales , Células de la Médula Ósea , Factores de Crecimiento Endotelial/administración & dosificación , Factores de Crecimiento Endotelial/sangre , Factores de Crecimiento Endotelial/metabolismo , Femenino , Vectores Genéticos , Movilización de Célula Madre Hematopoyética , Leucocitos/fisiología , Linfocinas/administración & dosificación , Linfocinas/sangre , Linfocinas/metabolismo , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Bazo/citología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
9.
Ann Oncol ; 20(3): 413-24, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19088170

RESUMEN

Angiogenesis, the growth of new blood vessels, requires dynamic expansion, assembly and stabilization of vascular endothelial cells in response to proangiogenic stimuli. Antiangiogenic strategies have become an important therapeutic modality for solid tumors. While many aspects of postnatal pathological angiogenesis have been extensively studied in the context of nonhematopoietic neoplasms, the precise role of these processes in lymphoma pathogenesis is under active investigation. Lymphoma growth and progression is potentiated by at least two distinct angiogenic mechanisms: autocrine stimulation of tumor cells via expression of vascular endothelial growth factor (VEGF) and VEGF receptors by lymphoma cells, as well as paracrine influences of proangiogenic tumor microenvironment on both local neovascular transformation and recruitment of circulating bone marrow-derived progenitors. Lymphoma-associated infiltrating host cells including hematopoietic monocytes, T cells and mesenchymal pericytes have increasingly been associated with the pathogenesis and prognosis of lymphoma, in part providing perivascular guidance and support to neoangiogenesis. Collectively, these distinct angiogenic mechanisms appear to be important therapeutic targets in selected non-Hodgkin's lymphoma (NHL) subtypes. Understanding these pathways has led to the introduction of antiangiogenic treatment strategies into the clinic where they are currently under assessment in several ongoing studies of NHL patients.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Linfoma no Hodgkin/tratamiento farmacológico , Neovascularización Patológica , Humanos , Linfoma no Hodgkin/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
J Cell Biol ; 153(6): 1277-86, 2001 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-11402070

RESUMEN

E-selectin plays a critical role in mediating tissue-specific homing of T cells into skin, and of primitive hematopoietic progenitor cells (HPCs) into bone marrow (BM). Though it is known that a glycoform of PSGL-1 (CLA) functions as the principal E-selectin ligand on human T lymphocytes, the E-selectin ligand(s) of human HPCs has not been identified. We used a shear-based adherence assay to analyze and define the E-selectin ligand activity of membrane proteins from human HPCs. Our data show that PSGL-1 expressed on human HPCs is an E-selectin ligand, and that HPCs also express a previously unrecognized E-selectin ligand, CD44. The E-selectin ligand activity of CD44 is conferred by the elaboration of sialylated, fucosylated binding determinants on N-glycans. This glycoform of CD44 is expressed on primitive CD34+ human HPCs, but not on more mature hematopoietic cells. Under physiologic flow conditions, this molecule mediates E-selectin-dependent rolling interactions over a wider shear range than that of PSGL-1, and promotes human HPC rolling interactions on E-selectin expressed on human BM endothelial cells. These findings offer new insights into the structural biology and physiology of CD44, and into the molecular basis of E-selectin-dependent adhesive interactions that direct homing of human HPC to BM.


Asunto(s)
Selectina E/metabolismo , Células Madre Hematopoyéticas/metabolismo , Receptores de Hialuranos/metabolismo , Animales , Western Blotting/métodos , Células CHO , Cricetinae , Selectina E/genética , Electroforesis en Gel de Poliacrilamida/métodos , Células HL-60 , Células Madre Hematopoyéticas/citología , Humanos , Receptores de Hialuranos/genética , Células K562 , Ligandos , Glicoproteínas de Membrana/metabolismo , Selectina-P/metabolismo , Dodecil Sulfato de Sodio
11.
J Clin Invest ; 105(1): 101-11, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10619866

RESUMEN

Stromal cell-derived factor-1 (SDF-1) provides a potent chemotactic stimulus for CD34(+) hematopoietic cells. We cultured mobilized peripheral blood (PB) and umbilical cord blood (CB) for up to 5 weeks and examined the migratory activity of cobblestone area-forming cells (CAFCs) and long-term culture-initiating cells (LTC-ICs) in a transwell assay. In this system, SDF-1 or MS-5 marrow stromal cells placed in the lower chamber induced transmembrane and transendothelial migration by 2- and 5-week-old CAFCs and LTC-ICs in 3 hours. Transmigration was blocked by preincubation of input CD34(+) cells with antibody to CXCR4. Transendothelial migration of CB CAFCs and LTC-ICs was higher than that of PB. We expanded CD34(+) cells from CB in serum-free medium with thrombopoietin, flk-2 ligand, and c-kit ligand, with or without IL-3 and found that CAFCs cultured in the absence of IL-3 had a chemotactic response equivalent to noncultured cells, even after 5 weeks. However, addition of IL-3 to the culture reduced this response by 20-50%. These data indicate that SDF-1 induces chemotaxis of primitive hematopoietic cells signaling through CXCR4 and that the chemoattraction could be downmodulated by culture ex vivo.


Asunto(s)
Quimiocinas CXC/farmacología , Quimiotaxis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Antígenos CD34/análisis , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12 , Endotelio Vascular/citología , Células Madre Hematopoyéticas/fisiología , Proteínas de la Membrana/farmacología , Ratones , Receptores CXCR4/análisis , Receptores CXCR4/fisiología , Factor de Células Madre/farmacología , Trombopoyetina/farmacología
12.
J Clin Invest ; 106(4): 511-21, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10953026

RESUMEN

Emerging data suggest that VEGF receptors are expressed by endothelial cells as well as hematopoietic stem cells. Therefore, we hypothesized that functional VEGF receptors may also be expressed in malignant counterparts of hematopoietic stem cells such as leukemias. We demonstrate that certain leukemias not only produce VEGF but also express functional VEGFR-2 in vivo and in vitro, resulting in the generation of an autocrine loop that may support leukemic cell survival and proliferation. Approximately 50% of freshly isolated leukemias expressed mRNA and protein for VEGFR-2. VEGF(165) induced phosphorylation of VEGFR-2 and increased proliferation of leukemic cells, demonstrating these receptors were functional. VEGF(165) also induced the expression of MMP-9 by leukemic cells and promoted their migration through reconstituted basement membrane. The neutralizing mAb IMC-1C11, specific to human VEGFR-2, inhibited leukemic cell survival in vitro and blocked VEGF(165)-mediated proliferation of leukemic cells and VEGF-induced leukemic cell migration. Xenotransplantation of primary leukemias and leukemic cell lines into immunocompromised nonobese diabetic mice resulted in significant elevation of human, but not murine, VEGF in plasma and death of inoculated mice within 3 weeks. Injection of IMC-1C11 inhibited proliferation of xenotransplanted human leukemias and significantly increased the survival of inoculated mice. Interruption of signaling by VEGFRs, particularly VEGFR-2, may provide a novel strategy for inhibiting leukemic cell proliferation.


Asunto(s)
Leucemia/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Secuencia de Bases , División Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Cartilla de ADN/genética , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Expresión Génica , Supervivencia de Injerto , Humanos , Leucemia/genética , Leucemia/patología , Linfocinas/genética , Linfocinas/metabolismo , Linfocinas/farmacología , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos NOD , Trasplante de Neoplasias , Células Neoplásicas Circulantes , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Transducción de Señal , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Ann N Y Acad Sci ; 1106: 175-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17395736

RESUMEN

Megakaryocytes and platelets have been known to secrete angiogenic growth factors for a long time. However, there is little in vivo data on the regulation of angiogenesis by thrombopoietic cells. Both megakaryocytes and platelets are known to carry and release a multitude of both pro- and antiangiogenic mediators. Thus, it remained unknown how the "angiogenic phenotype" of thrombopoietic cells would be determined. Our group established that platelets contribute to angiogenesis as carriers of SDF-1, which is released by platelets in response to stimulation with hematopoietic cytokines. Indeed, even the action of VEGF-A seems to be mediated in part by the release of SDF-1 from stimulated platelets, thereby attracting proangiogenic hematopoietic cells. Moreover, the analysis of murine plasma and serum showed that similar to VEGF-A, SDF-1 is almost exclusively derived from platelets, and only trace amounts are detectable in platelet poor plasma. Because tumor patients' platelets have been shown to contain lower amounts of thrombospondin (Tsp), we generated Tsp-1 and Tsp-2 double knockout mice by crossing the single knockout lines. Interestingly, megakaryocytes and platelets derived from these mice confer a proangiogenic phenotype both in the bone marrow and in reperfusion of ischemic hindlimbs, thereby verifying the hypothesis of pro- and antiangiogenic platelet constituents "in balance."


Asunto(s)
Plaquetas/citología , Células de la Médula Ósea/citología , Megacariocitos/citología , Animales , Plaquetas/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Endotelio Vascular/citología , Humanos , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Neovascularización Patológica , Fenotipo
14.
Leukemia ; 20(6): 979-86, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16598308

RESUMEN

Fibroblast growth factors (FGFs) are important regulators of hematopoiesis and have been implicated in the tumorigenesis of solid tumors. Recent evidence suggests that FGF signaling through FGF receptors (FGFRs) may play a role in the proliferation of subsets of acute myeloid leukemias (AMLs). However, the precise mechanism and specific FGF receptors that support leukemic cell growth are not known. We show that FGF-2, through activation of FGFR1beta signaling, promotes survival, proliferation and migration of AML cells. Stimulation of FGFR1beta results in phosphoinositide 3-kinase (PI3-K)/Akt activation and inhibits chemotherapy-induced apoptosis of leukemic cells. Neutralizing FGFR1-specific antibody abrogates the physiologic and chemoprotective effects of FGF-2/FGFR1beta signaling and inhibits tumor growth in mice xenotransplanted with human AML. These data suggest that activation of FGF-2/FGFR1beta supports progression and chemoresistance in subsets of AML. Therefore, FGFR1 targeting may be of therapeutic benefit in subsets of AML.


Asunto(s)
Movimiento Celular , Resistencia a Antineoplásicos , Leucemia Mieloide/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Enfermedad Aguda , Anciano de 80 o más Años , Proteína Relacionada con Agouti , Animales , Anticuerpos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/genética , Masculino , Ratones , Ratones SCID , Fosforilación , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Circ Res ; 88(9): 903-10, 2001 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-11348999

RESUMEN

Intravascular introduction of replication-deficient adenoviral vectors (Advectors) provides an ideal model of delivery of transgenes for the treatment of various vascular abnormalities. On the basis of the knowledge that Advectors can induce inflammatory responses after intravascular administration, we speculated that cellular activation by Advector infection could directly modulate the endothelial cell (EC) adhesion molecule/chemokine expression repertoire. Infection of human umbilical vein ECs or bone marrow microvascular ECs with an E1(-)E4(+) Advector resulted in the upregulation of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and CD34, but not E-selectin, P-selectin, CD36, CD13, CD44, HLA-DR or PECAM. Upregulation of ICAM-1, VCAM-1, and CD34 was apparent 12 hours after infection and persisted for weeks after infection. Selective induction of adhesion molecules was mediated by the presence of the E4 gene in the Advector, because infection of ECs with an E1(-)E4(-) Advector had no effect on adhesion molecule expression. ECs infected with E1(-)E4(+) Advector, but not those infected with E1(-)E4(-) Advector, supported the adhesion of leukocytes. Monoclonal antibodies to ICAM-1 and VCAM-1 inhibited adhesion of leukocytes to E1(-)E4(+)-infected ECS: Infection of the ECs with E1(-)E4(+) Advector, but not E1(-)E4(-) Advector, resulted in downregulation of expression of chemocytokines, including interleukin-8, MCP-1, RANTES, and GM-CSF. Nonetheless, a large number of leukocytes migrated through ECs infected with E1(-)E4(+), but not those infected with E1(-)E4(l-), in response to exogenous chemokines. These results demonstrate that infection of ECs with E1(-)E4(+) Advectors, but not E1(-)E4(-) Advectors, may directly augment inflammatory responses by upregulating expression of adhesion molecules and enhancing migration through Advector-infected ECs and suggest that E1(-)E4(-) Advectors may be a better choice for gene-transfer strategies directed to the ECS:


Asunto(s)
Proteínas E1 de Adenovirus/genética , Proteínas E4 de Adenovirus/genética , Endotelio Vascular/metabolismo , Leucocitos/metabolismo , Proteínas/metabolismo , Adenoviridae/genética , Antígenos CD34/genética , Antígenos CD34/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación hacia Abajo , Endotelio Vascular/citología , Regulación de la Expresión Génica , Vectores Genéticos/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/citología , Proteínas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Circ Res ; 88(2): 167-74, 2001 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-11157668

RESUMEN

Bone marrow (BM)-derived circulating endothelial precursor cells (CEPs) are thought to play a role in postnatal angiogenesis. Emerging evidence suggests that angiogenic stress of vascular trauma may induce mobilization of CEPs to the peripheral circulation. In this regard, we studied the kinetics of CEP mobilization in two groups of patients who experienced acute vascular insult secondary to burns or coronary artery bypass grafting (CABG). In both burn and CABG patients, there was a consistent, rapid increase in the number of CEPs, determined by their surface expression pattern of vascular endothelial growth factor receptor 2 (VEGFR2), vascular endothelial cadherin (VE-cadherin), and AC133. Within the first 6 to 12 hours after injury, the percentage of CEPs in the peripheral blood of burn or CABG patients increased almost 50-fold, returning to basal levels within 48 to 72 hours. Mobilized cells also formed late-outgrowth endothelial colonies (CFU-ECs) in culture, indicating that a small, but significant, number of circulating endothelial cells were BM-derived CEPs. In parallel to the mobilization of CEPs, there was also a rapid elevation of VEGF plasma levels. Maximum VEGF levels were detected within 6 to 12 hours of vascular trauma and decreased to baseline levels after 48 to 72 hours. Acute elevation of VEGF in the mice plasma resulted in a similar kinetics of mobilization of VEGFR2(+) cells. On the basis of these results, we propose that vascular trauma may induce release of chemokines, such as VEGF, that promotes rapid mobilization of CEPs to the peripheral circulation. Strategies to improve the mobilization and incorporation of CEPs may contribute to the acceleration of vascularization of the injured vascular tissue.


Asunto(s)
Vasos Sanguíneos/metabolismo , Endotelio Vascular/metabolismo , Glicoproteínas/metabolismo , Péptidos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Células Madre/metabolismo , Antígeno AC133 , Animales , Antígenos CD , Quemaduras/sangre , Cadherinas/genética , Cadherinas/metabolismo , Recuento de Células , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Puente de Arteria Coronaria , Factores de Crecimiento Endotelial/sangre , Endotelio Vascular/citología , Citometría de Flujo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Antígeno Lewis X/metabolismo , Linfocinas/sangre , Antígeno de Macrófago-1/metabolismo , Ratones , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Factor de von Willebrand/metabolismo
17.
Oncogene ; 20(58): 8334-41, 2001 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-11840326

RESUMEN

Since the identification of the Id proteins over a decade ago, a great many cell cycle and cell fate decisions have been shown to be under the control of these proteins as described in other sections of this review issue. Perhaps the most unsuspected activity of this class of proteins has been their essential role in angiogenesis, both in the forebrain during development and during the growth and metastasis of tumors in adults. This section of the review issue will focus on the key observations which have led to these conclusions, speculations about potential mechanisms and the outlook for potential therapeutic interventions.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Familia de Multigenes , Neovascularización Fisiológica/fisiología , Proteínas Represoras , Factores de Transcripción/fisiología , Animales , Vasos Sanguíneos/lesiones , Células de la Médula Ósea/citología , Diferenciación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Perros , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/citología , Secuencias Hélice-Asa-Hélice , Células Madre Hematopoyéticas/citología , Humanos , Proteína 1 Inhibidora de la Diferenciación , Proteína 2 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Linfocinas/fisiología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Células Madre/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Leukemia ; 17(3): 604-11, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12646950

RESUMEN

Vascular endothelial growth factor (VEGF) and its receptors (VEGFR) have been implicated in promoting solid tumor growth and metastasis via stimulating tumor-associated angiogenesis. We recently showed that certain 'liquid' tumors such as leukemia not only produce VEGF, but also express functional VEGFR, resulting in an autocrine loop for tumor growth and propagation. A chimeric anti-VEGFR2 (or kinase insert domain-containing receptor, KDR) antibody, IMC-1C11, was shown to be able to inhibit VEGF-induced proliferation of human leukemia cells in vitro, and to prolong survival of nonobese diabetic-severe combined immune deficient (NOD-SCID) mice inoculated with human leukemia cells. Here we produced two fully human anti-KDR antibodies (IgG1), IMC-2C6 and IMC-1121, from Fab fragments originally isolated from a large antibody phage display library. These antibodies bind specifically to KDR with high affinities: 50 and 200 pM for IMC-1121 and IMC-2C6, respectively, as compared to 270 pM for IMC-1C11. Like IMC-1C11, both human antibodies block VEGF/KDR interaction with an IC(50) of approximately 1 nM, but IMC-1121 is a more potent inhibitor to VEGF-stimulated proliferation of human endothelial cells. These anti-KDR antibodies strongly inhibited VEGF-induced migration of human leukemia cells in vitro, and when administered in vivo, significantly prolonged survival of NOD-SCID mice inoculated with human leukemia cells. It is noteworthy that the mice treated with antibody of the highest affinity, IMC-1121, survived the longest period of time, followed by mice treated with IMC-2C6 and IMC-1C11. Taken together, our data suggest that anti-KDR antibodies may have broad applications in the treatment of both solid tumors and leukemia. It further underscores the efforts to identify antibodies of high affinity for enhanced antiangiogenic and antitumor activities.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Leucemia Experimental/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Afinidad de Anticuerpos , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Células HL-60 , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Leucemia Experimental/mortalidad , Linfocinas/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión , Tasa de Supervivencia , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
19.
Exp Hematol ; 28(6): 700-6, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10880756

RESUMEN

In the hematopoietic microenvironment, bone marrow endothelial cells may play an important role in trafficking and maintenance of progenitor and stem cells due to adhesive interactions and paracrine secretion of hematopoietic growth factors. However, it is unknown whether progenitors in turn modulate endothelial proliferation and function. We analyzed mRNA expression (Northern blot) and release of vascular endothelial growth factor-A (VEGF-A), which specifically acts on endothelial cells, by cytokine-stimulated peripheral blood-derived CD34+ hematopoietic progenitor cells. While unstimulated CD34+ cells expressed VEGF-A mRNA weakly without cytokine release in vitro, incubation for 24 hours with a single cytokine (e.g., kit ligand [KL]) resulted in increased VEGF-A mRNA expression and significant secretion of VEGF-A into the supernatant. The amount of VEGF released was substantially augmented by incubation with a combination of cytokines (e.g., KL, IL-3, GM-CSF, G-CSF), or by exposure to hematopoietic cytokines for a longer time period. In addition, we show that VEGF induced the release of hematopoietic growth factors (GM-CSF) by bone marrow endothelial cells and that in vitro stromal cell-derived factor-1 (SDF-1) driven transendothelial progenitor cell migration was increased by the presence of VEGF, which might be due to pore formation (increased endothelial fenestration). In vivo, release of VEGF by progenitor cells may result in a paracrine loop supporting proliferation of both endothelium and progenitors and may facilitate transendothelial migration during cytokine-induced progenitor cell mobilization.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Hematopoyesis , Factores de Crecimiento de Célula Hematopoyética/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Antígenos CD34/análisis , Northern Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Permeabilidad Capilar , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Factores de Crecimiento Endotelial/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular
20.
Exp Hematol ; 29(12): 1417-24, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11750100

RESUMEN

OBJECTIVE: The mechanism for anagrelide's potent platelet lowering activity in human subjects is not well defined. Studies related to anagrelide function have been hampered by its lack of activity in nonhuman primates and water insolubility. In an effort to define the mechanism whereby anagrelide exerts its therapeutic effect, we identified a water-soluble metabolite (anagrelide.met). The availability of anagrelide.met allowed, for the first time, parallel in vitro and in vivo animal studies centered on the mechanisms by which anagrelide lowers platelet levels. MATERIALS AND METHODS: The effects of anagrelide.met on proliferation and maturation of mega-karyocytes (MKs) as well as platelet production were studied both in vitro and in vivo. RESULTS: Anagrelide.met is capable of blocking in vitro MK migration by 20% to 40%. At 100 ng/mL, anagrelide.met selectively blocked in vitro MK maturation, resulting in a 50% decrease in the total number of CD41a(+) MKs, corresponding with a 30% decrease in MK ploidy by day 10 and a 60% decrease by day 20. Daily intraperitoneal injections of anagrelide.met 100 microg into BALB/c mice was sufficient to significantly decrease platelet counts within 24 to 48 hours, stabilizing to 40 to 50% of normal levels by day 5. This was associated with a 45% decrease in the number of developing MKs and an increase in thrombopoietin levels. Anagrelide.met did not alter WBC counts, hematocrit, or bleeding time, or lead to any apparent signs of toxicity. Furthermore, unlike the parent anagrelide compound, anagrelide.met did not inhibit ADP-induced platelet aggregation even at high concentrations (10 microg/mL). CONCLUSIONS: We describe a cross-species reactive anagrelide metabolite that selectively inhibits MK maturation and migration, lowering platelet levels without influencing platelet aggregation.


Asunto(s)
Megacariocitos/patología , Trombocitopenia/fisiopatología , Acetilcolinesterasa/análisis , Animales , Antígenos CD34/análisis , Biomarcadores/análisis , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Sangre Fetal/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Recuento de Plaquetas , Ploidias , Quinazolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA