Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Br J Haematol ; 203(4): 504-506, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37803499

RESUMEN

Invasive fungal infections (IFIs), mainly due to pulmonary aspergillosis, are considered a serious complication in acute leukaemia, with an unfavourable impact on patient. In this well-conducted retrospective study, Reynolds et al. suggest that the use of posaconazole prophylaxis in association with venetoclax plus hypomethylating agents or chemotherapy leads to a reduction of IFI incidence. Therapeutic drug monitoring of posaconazole levels is suggested, even if no correlation with IFI risk has been demonstrated. Commentary on: Reynolds et al. Invasive fungal infection following venetoclax and posaconazole co-administration. Br J Haematol 2023;203:593-598.


Asunto(s)
Infecciones Fúngicas Invasoras , Leucemia Mieloide Aguda , Humanos , Antifúngicos/uso terapéutico , Estudios Retrospectivos , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/complicaciones , Infecciones Fúngicas Invasoras/tratamiento farmacológico
2.
Semin Hematol ; 60(2): 118-124, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37099031

RESUMEN

Recent advances in the understanding of Waldenström macroglobulinemia (WM) biology have impacted the development of effective novel agents and improved our knowledge of how the genomic background of WM may influence selection of therapy. Consensus Panel 7 (CP7) of the 11th International Workshop on WM was convened to examine the current generation of completed and ongoing clinical trials involving novel agents, consider updated data on WM genomics, and make recommendations on the design and prioritization of future clinical trials. CP7 considers limited duration and novel-novel agent combinations to be the priority for the next generation of clinical trials. Evaluation of MYD88, CXCR4 and TP53 at baseline in the context of clinical trials is crucial. The common chemoimmunotherapy backbones, bendamustine-rituximab (BR) and dexamethasone, rituximab and cyclophosphamide (DRC), may be considered standard-of-care for the frontline comparative studies. Key unanswered questions include the definition of frailty in WM; the importance of attaining a very good partial response or better (≥VGPR), within stipulated time frame, in determining survival outcomes; and the optimal treatment of WM populations with special needs.


Asunto(s)
Macroglobulinemia de Waldenström , Humanos , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/genética , Rituximab/uso terapéutico , Consenso , Ciclofosfamida/uso terapéutico , Clorhidrato de Bendamustina/uso terapéutico
3.
Leukemia ; 31(11): 2426-2434, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28344315

RESUMEN

The extracellular matrix (ECM) is a major component of the tumor microenvironment, contributing to the regulation of cell survival, proliferation, differentiation and metastasis. In multiple myeloma (MM), interactions between MM cells and the bone marrow (BM) microenvironment, including the BM ECM, are critical to the pathogenesis of the disease and the development of drug resistance. Nevertheless, composition of the ECM in MM and its role in supporting MM pathogenesis has not been reported. We have applied a novel proteomic-based strategy and defined the BM ECM composition in patients with monoclonal gammopathy of undetermined significance (MGUS), newly diagnosed and relapsed MM compared with healthy donor-derived BM ECM. In this study, we show that the tumor ECM is remodeled at the mRNA and protein levels in MGUS and MM to allow development of a permissive microenvironment. We further demonstrate that two ECM-affiliated proteins, ANXA2 and LGALS1, are more abundant in MM and high expression is associated with a decreased overall survival. This study points to the importance of ECM remodeling in MM and provides a novel proteomic pipeline for interrogating the role of the ECM in cancers with BM tropism.


Asunto(s)
Médula Ósea/metabolismo , Matriz Extracelular/metabolismo , Mieloma Múltiple/metabolismo , Proteoma , Anexina A2/metabolismo , Estudios de Casos y Controles , Galectina 1/metabolismo , Perfilación de la Expresión Génica , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Análisis de Supervivencia , Microambiente Tumoral
4.
Leukemia ; 31(4): 853-860, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27773931

RESUMEN

MYC is a major oncogenic driver of multiple myeloma (MM) and yet almost no therapeutic agents exist that target MYC in MM. Here we report that the let-7 biogenesis inhibitor LIN28B correlates with MYC expression in MM and is associated with adverse outcome. We also demonstrate that the LIN28B/let-7 axis modulates the expression of MYC, itself a let-7 target. Further, perturbation of the axis regulates the proliferation of MM cells in vivo in a xenograft tumor model. RNA-sequencing and gene set enrichment analyses of CRISPR-engineered cells further suggest that the LIN28/let-7 axis regulates MYC and cell cycle pathways in MM. We provide proof of principle for therapeutic regulation of MYC through let-7 with an LNA-GapmeR (locked nucleic acid-GapmeR) containing a let-7b mimic in vivo, demonstrating that high levels of let-7 expression repress tumor growth by regulating MYC expression. These findings reveal a novel mechanism of therapeutic targeting of MYC through the LIN28B/let-7 axis in MM that may impact other MYC-dependent cancers as well.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Mieloma Múltiple/genética , Interferencia de ARN , Proteínas de Unión al ARN/genética , Animales , Estudios de Casos y Controles , Ciclo Celular/genética , Línea Celular Tumoral , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes myc , Xenoinjertos , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Pronóstico , Proteínas de Unión al ARN/metabolismo
5.
Leukemia ; 30(5): 1103-15, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26859080

RESUMEN

The role of endothelial progenitor cell (EPC)-mediated vasculogenesis in hematological malignancies is not well explored. Here, we showed that EPCs are mobilized from the bone marrow (BM) to the peripheral blood at early stages of multiple myeloma (MM); and recruited to MM cell-colonized BM niches. Using EPC-defective ID1+/- ID3-/- mice, we found that MM tumor progression is dependent on EPC trafficking. By performing RNA-sequencing studies, we confirmed that endothelial cells can enhance proliferation and favor cell-cycle progression only in MM clones that are smoldering-like and have dependency on endothelial cells for tumor growth. We further confirmed that angiogenic dependency occurs early and not late during tumor progression in MM. By using a VEGFR2 antibody with anti-vasculogenic activity, we demonstrated that early targeting of EPCs delays tumor progression, while using the same agent at late stages of tumor progression is ineffective. Thus, although there is significant angiogenesis in myeloma, the dependency of the tumor cells on EPCs and vasculogenesis may actually precede this step. Manipulating vasculogenesis at an early stage of disease may be examined in clinical trials in patients with smoldering MM, and other hematological malignancies with precursor conditions.


Asunto(s)
Mieloma Múltiple/patología , Neovascularización Patológica/tratamiento farmacológico , Animales , Anticuerpos/uso terapéutico , Médula Ósea , Movimiento Celular , Células Clonales/patología , Progresión de la Enfermedad , Células Endoteliales/patología , Ratones , Mieloma Múltiple/irrigación sanguínea , Mieloma Múltiple/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Prevención Secundaria , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
6.
Leukemia ; 29(12): 2338-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26139427

RESUMEN

We examined the combination of the mammalian target of rapamycin inhibitor everolimus with bortezomib and rituximab in patients with relapsed/refractory Waldenstrom macroglobulinemia (WM) in a phase I/II study. All patients received six cycles of the combination of everolimus/rituximab or everolimus/bortezomib/rituximab followed by maintenance with everolimus until progression. Forty-six patients were treated; 98% received prior rituximab and 57% received prior bortezomib. No dose-limiting toxicities were observed in the phase I. The most common treatment-related toxicities of all grades were fatigue (63%), anemia (54%), leucopenia (52%), neutropenia (48%) and diarrhea (43%). Thirty-six (78%) of the 46 patients received full dose therapy (FDT) of the three drugs. Of these 36, 2 (6%) had complete response (90% confidence interval (CI): 1-16). In all, 32/36 (89%) of patients experienced at least a minimal response (90% CI: 76-96%). The observed partial response or better response rate was 19/36 (53, 90 CI: 38-67%). For the 36 FDT patients, the median progression-free survival was 21 months (95% CI: 12-not estimable). In summary, this study demonstrates that the combination of everolimus, bortezomib and rituximab is well tolerated and achieved 89% response rate even in patients previously treated, making it a possible model of non-chemotherapeutic-based combination therapy in WM.


Asunto(s)
Macroglobulinemia de Waldenström/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Bortezomib/administración & dosificación , Bortezomib/efectos adversos , Quimioterapia Combinada , Everolimus/administración & dosificación , Everolimus/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Factor 88 de Diferenciación Mieloide/genética , Receptores CXCR4/genética , Recurrencia , Rituximab/administración & dosificación , Rituximab/efectos adversos
8.
J Biomed Biotechnol ; 2012: 157496, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093834

RESUMEN

Substantial advances have been made in understanding the biology of multiple myeloma (MM) through the study of the bone marrow (BM) microenvironment. Indeed, the BM niche appears to play an important role in differentiation, migration, proliferation, survival, and drug resistance of the malignant plasma cells. The BM niche is composed of a cellular compartment (stromal cells, osteoblasts, osteoclasts, endothelial cells, and immune cells) and a noncellular compartment including the extracellular matrix (ECM) and the liquid milieu (cytokines, growth factors, and chemokines). In this paper we discuss how the interaction between the malignant plasma cell and the BM microenvironment allowed myeloma progression through cell homing and the new concept of premetastatic niche.


Asunto(s)
Médula Ósea/inmunología , Citocinas/inmunología , Modelos Inmunológicos , Mieloma Múltiple/inmunología , Animales , Humanos
9.
Curr Cancer Drug Targets ; 11(9): 1025-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21933107

RESUMEN

Bone marrow microenvironment has been shown to play a crucial role in supporting the pathogenesis and the progression of several B-cell malignancies, including Waldenstrom's Macroglobulinemia (WM). Among the different cell types within the bone marrow milieu, endothelial cells have been proven to support WM cells growth. Based on the understanding of bone marrow neo-angiogenesis in plasma cell dyscrasias, a number of anti-angiogenic molecules are now available for the treatment of these diseases. Indeed, anti-angiogenic drugs, such as proteasome-, proteins kinase-C (PKC)-, phosphatidylinositol 3-kinase/mammalian target of rapamycin (mTOR)-, and histone deacetylase (HDAC)- inhibitors are now available, playing a key role in the treatment of WM both in the preclinical settings and as part of clinical trials.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/patología , Inhibidores de la Angiogénesis/farmacología , Médula Ósea/metabolismo , Médula Ósea/patología , Ácidos Borónicos/uso terapéutico , Bortezomib , Células Endoteliales/patología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Indoles/farmacología , Indoles/uso terapéutico , Panobinostat , Inhibidores de Proteasas , Inhibidores de Proteasoma , Proteína Quinasa C/antagonistas & inhibidores , Pirazinas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Microambiente Tumoral
10.
Curr Cancer Drug Targets ; 11(3): 254-84, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21247382

RESUMEN

The proteasome has emerged as an important clinically relevant target for the treatment of hematologic malignancies. Since the Food and Drug Administration approved the first-in-class proteasome inhibitor bortezomib (Velcade) for the treatment of relapsed/refractory multiple myeloma (MM) and mantle cell lymphoma, it has become clear that new inhibitors are needed that have a better therapeutic ratio, can overcome inherent and acquired bortezomib resistance and exhibit broader anti-cancer activities. Marizomib (NPI-0052; salinosporamide A) is a structurally and pharmacologically unique ß-lactone-γ-lactam proteasome inhibitor that may fulfill these unmet needs. The potent and sustained inhibition of all three proteolytic activities of the proteasome by marizomib has inspired extensive preclinical evaluation in a variety of hematologic and solid tumor models, where it is efficacious as a single agent and in combination with biologics, chemotherapeutics and targeted therapeutic agents. Specifically, marizomib has been evaluated in models for multiple myeloma, mantle cell lymphoma, Waldenstrom's macroglobulinemia, chronic and acute lymphocytic leukemia, as well as glioma, colorectal and pancreatic cancer models, and has exhibited synergistic activities in tumor models in combination with bortezomib, the immunomodulatory agent lenalidomide (Revlimid), and various histone deacetylase inhibitors. These and other studies provided the framework for ongoing clinical trials in patients with MM, lymphomas, leukemias and solid tumors, including those who have failed bortezomib treatment, as well as in patients with diagnoses where other proteasome inhibitors have not demonstrated significant efficacy. This review captures the remarkable translational studies and contributions from many collaborators that have advanced marizomib from seabed to bench to bedside.


Asunto(s)
Antineoplásicos/uso terapéutico , Lactonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteasoma , Pirroles/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Humanos , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo
11.
Bone Marrow Transplant ; 44(11): 729-37, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19398965

RESUMEN

The role of different cytokines and cells of immune system in the pathogenesis of chronic GVHD (cGVHD) is still controversial. Earlier studies, which were either retrospective or analysed one or a few factors, did not show unequivocal results. We prospectively evaluated cytokine levels and lymphocyte subsets in 30 patients who underwent Allo-SCT to investigate their possible correlation with cGVHD. Levels of IL-4, IL-6, IL-10, IFN-gamma, tumour necrosis factor-alpha (TNF-alpha) and its soluble receptors were assessed by ELISA in 30 patients at different times after SCT. Lymphocyte subsets were evaluated by flow cytometry in peripheral blood at the same times as cytokines. A multivariate analysis was performed using principal component analysis and multi-factor ANOVA (analysis of variance). Eighteen patients developed cGVHD at a median time of 6 months (range, 5-9) after SCT. In multivariate analysis, we observed a correlation between cGVHD and clusters of cytokines and lymphocyte subsets from the third to the sixth month after SCT. These clusters changed their composition over time, but they constantly included natural killer (NK) and CD152+ T cells as negative predictors of cGVHD. TNF-alpha prevailed among other cytokines before the onset of cGVHD. This prevalence could be related partly to the defect of immunoregulatory cells.


Asunto(s)
Citocinas/inmunología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre de Sangre Periférica/métodos , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Células Th2/inmunología , Adulto , Anciano , Enfermedad Crónica , Citocinas/sangre , Femenino , Enfermedad Injerto contra Huésped/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Acondicionamiento Pretrasplante/métodos , Adulto Joven
12.
Leukemia ; 21(1): 44-52, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16990761

RESUMEN

Spontaneously arising tumor cells are not usually angiogenic at first. The phenotypic switch to angiogenesis is usually accomplished by a substet that induces new capillaries that then converge toward the tumor. The switch clearly involves more than simple upregulation of angiogenic activity and is thought to be the result of a net balance of positive and negative regulators. Tumor growth is although to require disruption of this balance and hence this switch must turned on for cancer progression. Progenitor endothelial cells, the crosstalk between angiogenic factors and their receptors and the interaction between vasculogenesis and lymphangiogenesis are all factors that may contribute to the switch. Its promotion is also the outcome of genetic instability resulting in the emergence of tumor cell lines. This review describes the history of the angiogenic switch illustrated in the literature and with particular reference to the three transgenic mouse models, namely RIP1-TAG2, keratin-14 (K14) (human papilloma virus) HPV16 and papilloma virus, used for stage-specific assessment of the effects of antiangiogenic and antitumorigenic agents.


Asunto(s)
Neoplasias/irrigación sanguínea , Neovascularización Patológica , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Células de la Médula Ósea/patología , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Humanos , Queratina-14/genética , Queratina-14/metabolismo , Ratones , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Transducción de Señal , Células Madre/patología , Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Curr Pharm Biotechnol ; 7(6): 441-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17168660

RESUMEN

The ubiquitin-proteasome pathway (UPP) is the major non-lysosomal proteolytic system in the cytosol and nucleus of all eukaryotic cells. Bortezomib (also known as PS-341 and Velcade) is a proteasome inhibitor, a novel class of cancer therapies. Bortezomib blocks multi-ubiquitinated protein degradation by inhibiting 26S proteasome activity, including regulating cell cycle, anti-apoptosis, and inflammation, as well as immune surveillance. In multiple myeloma (MM) cells, bortezomib directly induces cell stress response followed by activation of c-Jun NH(2) terminal kinase (JNK)/stress-activated protein kinase (SAPK), and triggers caspase-dependent apoptosis of tumor cells. Recent clinical studies demonstrated that bortezomib had remarkable anti-tumor activity in refractory and relapsed MM, providing the basis to approval by FDA. Its anti-tumor activities earlier in the course, in combination therapies, and in other malignancies is ongoing.


Asunto(s)
Ácidos Borónicos/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Pirazinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Animales , Antineoplásicos/administración & dosificación , Bortezomib , Ensayos Clínicos como Asunto , Humanos
14.
Neuroscience ; 140(3): 835-48, 2006 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-16650610

RESUMEN

Brain edema and severe alterations of the glial and endothelial cells have recently been demonstrated in the dystrophin-deficient mdx mouse, an experimental model of Duchenne muscular dystrophy, and an increase in microvessel density in patients affected by Duchenne muscular dystrophy has also been shown. In order to further elucidate the mechanisms underlying the angiogenetic processes occurring in Duchenne muscular dystrophy, in this study we analyzed matrix-metalloproteinase-2 and -9 expression in the brain of 20-month-old mdx and control mice by means of immunohistochemistry, in situ hybridization, immunoblotting and gelatin zymography. Moreover, we studied vascular endothelial growth factor expression by means of Western blot and immunohistochemistry, and by dual immunofluorescence using anti-vascular endothelial growth factor and anti matrix-metalloproteinase-2 and-9 antibodies. Ultrastructural features of the brain choroidal plexuses were evaluated by electron microscopy. Spatial relationships between endothelium and astrocyte processes were studied by confocal laser microscopy, using an anti-CD31 antibody as a marker of endothelial cells, and anti-glial fibrillary acidic protein (GFAP) as a marker of glial cells. The results demonstrate that high expression of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 protein content occurs in mdx brain and in choroidal plexuses where, by in situ hybridization, matrix-metalloproteinase-2 and matrix-metalloproteinase-9 mRNA was localized in the epithelial cells. Moreover, matrix-metalloproteinase-2 mRNA was found in both mdx perivascular astrocytes and blood vessels, while matrix-metalloproteinase-9 mRNA was localized in mdx vessels. Through zymography, increased expression of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 was found in mdx brain compared with the controls. These enhanced matrix-metalloproteinase levels in mdx mice were found to be associated with increased vascular endothelial growth factor expression, as determined by immunoblotting and immunocytochemistry and with ultrastructural alterations of the mdx choroidal epithelial cells and brain vessels, as previously reported [Nico B, Frigeri A, Nicchia GP, Corsi P, Ribatti D, Quondamatteo F, Herken R, Girolamo F, Marzullo A, Svelto M, Roncali L (2003) Severe alterations of endothelial and glial cells in the blood-brain barrier of dystrophic mdx mice. Glia 42:235-251]. Indeed, in the mdx epithelial cells of the plexuses, the apical microvilli were located on the lateral membranes, whereas in the controls they were uniformly distributed over the free ventricular surface. Moreover, by dual immunofluorescence, a colocalization of vascular endothelial growth factor and matrix-metalloproteinase-2 and matrix-metalloproteinase-9 was found in the ependymal and epithelial cells of plexuses in mdx mice and, under confocal laser microscopy, mdx CD-31 positive vessels were enveloped by less GFAP-positive astrocyte processes than the controls. Overall, these data point to a specific pathogenetic role of matrix-metalloproteinase-2 and matrix-metalloproteinase-9 in neurological dysfunctions associated with Duchenne muscular dystrophy.


Asunto(s)
Barrera Hematoencefálica/enzimología , Encéfalo/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Microcirculación/enzimología , Distrofia Muscular de Duchenne/enzimología , Animales , Astrocitos/enzimología , Astrocitos/patología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/patología , Encéfalo/fisiopatología , Plexo Coroideo/enzimología , Plexo Coroideo/patología , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Epéndimo/enzimología , Epéndimo/patología , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Microcirculación/patología , Microcirculación/fisiopatología , Microscopía Electrónica de Transmisión , Microvellosidades/enzimología , Microvellosidades/patología , Distrofia Muscular de Duchenne/fisiopatología , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Leukemia ; 19(7): 1253-61, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15858615

RESUMEN

Thalidomide alone or in combination with steroids has significant activity in multiple myeloma (MM). However, given its teratogenic potential, analogs have been synthesized, retaining the anti-MM activity without these side effects. We examined the anti-MM activity of two thalidomide analogs, CPS11 and CPS49. Direct cytotoxicity of the drugs on myeloma cell lines and patient myeloma cells was examined using thymidine uptake. Tumor cell apoptosis was evaluated by flow cytometry as well as Western blotting for caspase and PARP cleavage. Cellular signaling events were examined by immunoblotting for phosphorylated proteins. Both drugs inhibit proliferation of several MM cell lines sensitive and resistant to conventional therapies. They decrease secretion of IL-6, IGF, and VEGF by marrow stromal cells. Importantly, they inhibit proliferation of MM cells adherent to stromal cells. These drugs induce caspase-mediated apoptosis in MM cell lines, as well as patient MM cells. They inhibit the PI3K/Akt and JAK/STAT (signal transducers and activators of transcription) pathways in MM cells and are antiangiogenic in matrigel-based assays. CPS11 and CPS49 have potent antimyeloma activity and can overcome protective effects of the tumor microenvironment. They have potent antiangiogenic activity and direct effect on bone marrow stroma. These encouraging preclinical data provide the basis for further evaluation in the clinic.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Talidomida/farmacología , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , ADN/biosíntesis , ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Células del Estroma/efectos de los fármacos
16.
Clin Exp Allergy ; 34(11): 1660-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15544587

RESUMEN

The current wisdom is that tumours are endowed with an angiogenic capability and that their growth, invasion and metastasis are angiogenesis dependent. It is now well documented that neoplastic cells are influenced by their microenvironment and vice versa. The specific organ microenvironment determines the extent of cancer cell proliferation, angiogenesis, invasion and survival. Tumour cells are surrounded by an infiltrate of inflammatory cells, namely lymphocytes, neutrophils, macrophages and mast cells (MCs), which communicate via a complex network of intercellular signalling pathways, mediated by surface adhesion molecules, cytokines and their receptors. This review article summarizes: (i) the MC mediators involved in angiogenesis; (ii) the experimental evidence concerning the role played by MCs in angiogenesis; (iii) the list of solid and haematological tumours in which a close relationship between angiogenesis, tumour progression and MCs has been demonstrated; (iv) the circumstances in which MCs are a critical source of angiogenic factors in vivo, and in such cases, the signals that regulate their production and secretion that need to be determined as a prelude to the elaboration of new therapeutic strategies associated with MC presence and activation.


Asunto(s)
Mastocitos/fisiología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/patología , Inductores de la Angiogénesis/metabolismo , Animales , Progresión de la Enfermedad , Neoplasias Hematológicas/patología , Humanos , Neoplasias/patología
17.
Leukemia ; 17(10): 1961-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14513045
18.
Eur J Clin Invest ; 33(10): 891-6, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14511361

RESUMEN

Erythropoietin (Epo) is produced by the fetal liver and adult kidney and is an essential stimulator of erythropoiesis. It has, however, been shown to modulate host cellular signal transduction pathway to perform many other functions. New sites of Epo production have been found, such as the female reproductive organs and central nervous system. This review summarizes the involvement of Epo in the regulation of angiogenesis in both normal and pathological conditions.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Eritropoyetina/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Encéfalo/irrigación sanguínea , Femenino , Genitales Femeninos/irrigación sanguínea , Humanos , Neoplasias/irrigación sanguínea
20.
Eur J Clin Invest ; 32(3): 199-206, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895472

RESUMEN

BACKGROUND: Changes in angiogenesis and expression of extracellular matrix-degrading enzymes have been substantiated in tumour changeover and progression. METHODS: Tissues from 44 biopsies of stage III and IV ovarian endometriomas, and 10 biopsies of normal (control) endometrium were investigated immunohistochemically to count microvessels, and by in situ hybridization to assess the expression of mRNA of matrix metalloproteinase-2 (MMP-2) and MMP-9. Implants of the tissues were investigated in the chick embryo chorioallantoic membrane (CAM) assay to determine their angiogenic capacity. RESULTS: The endometriomas displayed significantly higher counts than normal endometria and the highest values were associated with the deepest invasion level (stage IV). Microvessels localized in close association with ectopic endometrial cells in the form of winding and arborized tubes, often dilated in microaneurysmatic segments. These were absent in normal endometrium. Expression of MMP-2 and MMP-9 mRNA, evaluated as percentages of positive biopsies and intensity of expression, was up-regulated in endometriomas and more pronounced in stage IV. MMP-2 and MMP-9 mRNA were also expressed by host stromal cells, including microvascular endothelial cells, fibroblasts and macrophages, whereas the control endometrium showed very little expression of MMP-2 mRNA in a few endothelial cells and no expression of MMP-9 mRNA. Implants from stage IV endometrioma induced a more intense vasoproliferative response than those from stage III, while no vasoproliferative response was induced by the normal endometrium. CONCLUSION: These data suggest that angiogenesis and degradation of extracellular matrix occur together in endometriosis and are more pronounced in stage IV, and that endometriosis cells and some host stromal cell populations co-operate in disease progression.


Asunto(s)
Endometriosis/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Neovascularización Patológica/metabolismo , Quistes Ováricos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Alantoides , Animales , Embrión de Pollo , Endometriosis/patología , Endometrio/enzimología , Endometrio/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Neovascularización Patológica/patología , Quistes Ováricos/patología , ARN Mensajero/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA