Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Haematologica ; 105(4): 1032-1041, 2020 04.
Article in English | MEDLINE | ID: mdl-31296574

ABSTRACT

CD38 is expressed in several types of non-Hodgkin lymphoma (NHL) and constitutes a promising target for antibody-based therapy. Daratumumab (Darzalex) is a first-in-class anti-CD38 antibody approved for the treatment of relapsed/refractory (R/R) multiple myeloma (MM). It has also demonstrated clinical activity in Waldenström macroglobulinaemia and amyloidosis. Here, we have evaluated the activity and mechanism of action of daratumumab in preclinical in vitro and in vivo models of mantle cell lymphoma (MCL), follicular lymphoma (FL) and diffuse large B-cell lymphoma (DLBCL), as monotherapy or in combination with standard chemo-immunotherapy. In vitro, daratumumab engages Fc-mediated cytotoxicity by antibody-dependent cell cytotoxicity and antibody-dependent cell phagocytosis in all lymphoma subtypes. In the presence of human serum, complement-dependent cell cytotoxicity was marginally engaged. We demonstrated by Selective Plane Illumination Microscopy that daratumumab fully penetrated a three-dimensional (3D) lymphoma organoid and decreased organoid volume. In vivo, daratumumab completely prevents tumor outgrowth in models of MCL and FL, and shows comparable activity to rituximab in a disseminated in vivo model of blastic MCL. Moreover, daratumumab improves overall survival (OS) in a mouse model of transformed CD20dim FL, where rituximab showed limited activity. Daratumumab potentiates the antitumor activity of CHOP and R-CHOP in MCL and FL xenografts. Furthermore, in a patient-derived DLBCL xenograft model, daratumumab anti-tumor activity was comparable to R-CHOP and the addition of daratumumab to either CHOP or R-CHOP led to full tumor regression. In summary, daratumumab constitutes a novel therapeutic opportunity in certain scenarios and these results warrant further clinical development.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphoma, Non-Hodgkin/therapy , Adult , B-Lymphocytes , Humans , Immunotherapy , Rituximab
2.
Int J Cancer ; 144(11): 2762-2773, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30468254

ABSTRACT

The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has been shown to be highly effective in patients with chronic lymphocytic leukemia (CLL) and is approved for CLL treatment. Unfortunately, resistance and intolerance to ibrutinib has been observed in several studies, opening the door for more specific BTK inhibitors. CC-292 (spebrutinib) is a BTK inhibitor with increased specificity for BTK and less inhibition of other kinases. Our in vitro studies showed that CC-292 potently inhibited B-cell receptor signaling, activation, proliferation and chemotaxis of CLL cells. In in vivo studies using the adoptive transfer TCL1 mouse model of CLL, CC-292 reduced tumor load and normalized tumor-associated expansion of T cells and monocytes, while not affecting T cell function. Importantly, the combination of CC-292 and bendamustine impaired CLL cell proliferation in vivo and enhanced the control of CLL progression. Our results demonstrate that CC-292 is a specific BTK inhibitor with promising performance in combination with bendamustine in CLL. Further clinical trials are warranted to investigate the therapeutic efficacy of this combination regimen.


Subject(s)
Acrylamides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bendamustine Hydrochloride/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Acrylamides/therapeutic use , Adult , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bendamustine Hydrochloride/therapeutic use , Bone Marrow/pathology , Disease Models, Animal , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Primary Cell Culture , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Pyrimidines/therapeutic use , Tumor Cells, Cultured
3.
Front Microbiol ; 9: 986, 2018.
Article in English | MEDLINE | ID: mdl-29867886

ABSTRACT

Pseudomonas aeruginosa is a major pathogenic bacterium in chronic infections and is a model organism for studying biofilms. P. aeruginosa is considered an aerobic bacterium, but in the presence of nitrate, it also grows in anaerobic conditions. Oxygen diffusion through the biofilm generates metabolic and genetic diversity in P. aeruginosa growth, such as in ribonucleotide reductase activity. These essential enzymes are necessary for DNA synthesis and repair. Oxygen availability determines the activity of the three-ribonucleotide reductase (RNR) classes. Class II and III RNRs are active in the absence of oxygen; however, class II RNRs, which are important in P. aeruginosa biofilm growth, require a vitamin B12 cofactor for their enzymatic activity. In this work, we elucidated the conditions in which class II RNRs are active due to vitamin B12 concentration constraints (biosynthesis or environmental availability). We demonstrated that increased vitamin B12 levels during aerobic, stationary and biofilm growth activate class II RNR activity. We also established that the cobN gene is essentially responsible for B12 biosynthesis under planktonic and biofilm growth. Our results unravel the mechanisms of dNTP synthesis by P. aeruginosa during biofilm growth, which appear to depend on the bacterial strain (laboratory-type or clinical isolate).

4.
Sci Rep ; 7(1): 17892, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263410

ABSTRACT

Ribonucleotide reductases (RNR) catalyze the last step of deoxyribonucleotide synthesis, and are therefore essential to DNA-based life. Three forms of RNR exist: classes I, II, and III. While eukaryotic cells use only class Ia RNR, bacteria can harbor any combination of classes, granting them adaptability. The opportunistic pathogen Pseudomonas aeruginosa surprisingly encodes all three classes, allowing it to thrive in different environments. Here we study an aspect of the complex RNR regulation whose molecular mechanism has never been elucidated, the well-described induction through oxidative stress, and link it to the AlgZR two-component system, the primary regulator of the mucoid phenotype. Through bioinformatics, we identify AlgR binding locations in RNR promoters, which we characterize functionally through EMSA and physically through AFM imaging. Gene reporter assays in different growth models are used to study the AlgZR-mediated control on the RNR network under various environmental conditions and physiological states. Thereby, we show that the two-component system AlgZR, which is crucial for bacterial conversion to the mucoid phenotype associated with chronic disease, controls the RNR network and directs how the DNA synthesis pathway is modulated in mucoid and non-mucoid biofilms, allowing it to respond to oxidative stress.


Subject(s)
Bacterial Proteins/genetics , Oxidative Stress/genetics , Pseudomonas aeruginosa/genetics , Ribonucleotide Reductases/genetics , Biofilms/growth & development , Computational Biology/methods , DNA/genetics , Gene Expression Regulation, Bacterial/genetics , Genes, Reporter/genetics , Phenotype , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics
5.
Sci Rep ; 7(1): 13350, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042684

ABSTRACT

Pseudomonas aeruginosa strain PAO1 has become the reference strain in many laboratories. One enzyme that is essential for its cell division is the ribonucleotide reductase (RNR) enzyme that supplies the deoxynucleotides required for DNA synthesis and repair. P. aeruginosa is one of the few microorganisms that encodes three different RNR classes (Ia, II and III) in its genome, enabling it to grow and adapt to diverse environmental conditions, including during infection. In this work, we demonstrate that a lack of RNR activity induces cell elongation in P. aeruginosa PAO1. Moreover, RNR gene expression during anaerobiosis differs among P. aeruginosa strains, with class III highly expressed in P. aeruginosa clinical isolates relative to the laboratory P. aeruginosa PAO1 strain. A single point mutation was identified in the P. aeruginosa PAO1 strain class III RNR promoter region that disrupts its anaerobic transcription by the Dnr regulator. An engineered strain that induces the class III RNR expression allows P. aeruginosa PAO1 anaerobic growth and increases its virulence to resemble that of clinical strains. Our results demonstrate that P. aeruginosa PAO1 is adapted to laboratory conditions and is not the best reference strain for anaerobic or infection studies.


Subject(s)
Point Mutation , Promoter Regions, Genetic , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Ribonucleotide Reductases/genetics , Base Sequence , Biofilms , Gene Expression Regulation, Bacterial , Genes, Reporter , Humans , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism
6.
Sci Rep ; 7(1): 13946, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066743

ABSTRACT

Mantle cell lymphoma (MCL) is a hematologic neoplasm characterised by the t(11;14)(q13;q32) translocation leading to aberrant cyclin D1 expression. The cell functions of cyclin D1 depend on its partners and/or subcellular distribution, resulting in different oncogenic properties. We observed the accumulation of cyclin D1 in the cytoplasm of a subset of MCL cell lines and primary cells. In primary cells, this cytoplasmic distribution was correlated with a more frequent blastoid phenotype. We performed immunoprecipitation assays and mass spectrometry on enriched cytosolic fractions from two cell lines. The cyclin D1 interactome was found to include several factors involved in adhesion, migration and invasion. We found that the accumulation of cyclin D1 in the cytoplasm was associated with higher levels of migration and invasiveness. We also showed that MCL cells with high cytoplasmic levels of cyclin D1 engrafted more rapidly into the bone marrow, spleen, and brain in immunodeficient mice. Both migration and invasion processes, both in vivo and in vitro, were counteracted by the exportin 1 inhibitor KPT-330, which retains cyclin D1 in the nucleus. Our data reveal a role of cytoplasmic cyclin D1 in the control of MCL cell migration and invasion, and as a true operator of MCL pathogenesis.


Subject(s)
Cell Movement , Cyclin D1/metabolism , Cytoplasm/metabolism , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Active Transport, Cell Nucleus , Adult , Aged , Aged, 80 and over , Animals , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Cytosol/metabolism , Female , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Proteomics
8.
Clin Cancer Res ; 23(6): 1493-1505, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-27637890

ABSTRACT

Purpose: To establish a proof-of-concept for the efficacy of the anti-CD38 antibody daratumumab in the poor prognosis CD38+ chronic lymphocytic leukemia (CLL) subtype.Experimental Design: The mechanism of action of daratumumab was assessed in CLL primary cells and cell lines using peripheral blood mononuclear cells to analyze antibody-dependent cell cytotoxicity (ADCC), murine and human macrophages to study antibody-dependent cell phagocytosis (ADCP), or human serum to analyze complement-dependent cytotoxicity (CDC). The effect of daratumumab on CLL cell migration and adhesion to extracellular matrix was characterized. Daratumumab activity was validated in two in vivo models.Results: Daratumumab demonstrated efficient lysis of patient-derived CLL cells and cell lines by ADCC in vitro and ADCP both in vitro and in vivo whereas exhibited negligible CDC in these cells. To demonstrate the therapeutic effect of daratumumab in CLL, we generated a disseminated CLL mouse model with the CD38+ MEC2 cell line and CLL patient-derived xenografts (CLL-PDX). Daratumumab significantly prolonged overall survival of MEC2 mice, completely eliminated cells from the infiltrated organs, and significantly reduced disease burden in the spleen of CLL-PDX. The effect of daratumumab on patient-derived CLL cell dissemination was demonstrated in vitro by its effect on CXCL12-induced migration and in vivo by interfering with CLL cell homing to spleen in NSG mice. Daratumumab also reduced adhesion of CLL cells to VCAM-1, accompanied by downregulation of the matrix metalloproteinase MMP9.Conclusions: These unique and substantial effects of daratumumab on CLL viability and dissemination support the investigation of its use in a clinical setting of CLL. Clin Cancer Res; 23(6); 1493-505. ©2016 AACR.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Antibodies, Monoclonal/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Tumor Microenvironment/drug effects , ADP-ribosyl Cyclase 1/immunology , Animals , Cell Line, Tumor , Cytophagocytosis/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Matrix Metalloproteinase 9/genetics , Mice , Xenograft Model Antitumor Assays
10.
Nature ; 535(7612): 382-7, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27443740

ABSTRACT

Cells deficient in the Brca1 and Brca2 genes have reduced capacity to repair DNA double-strand breaks by homologous recombination and consequently are hypersensitive to DNA-damaging agents, including cisplatin and poly(ADP-ribose) polymerase (PARP) inhibitors. Here we show that loss of the MLL3/4 complex protein, PTIP, protects Brca1/2-deficient cells from DNA damage and rescues the lethality of Brca2-deficient embryonic stem cells. However, PTIP deficiency does not restore homologous recombination activity at double-strand breaks. Instead, its absence inhibits the recruitment of the MRE11 nuclease to stalled replication forks, which in turn protects nascent DNA strands from extensive degradation. More generally, acquisition of PARP inhibitors and cisplatin resistance is associated with replication fork protection in Brca2-deficient tumour cells that do not develop Brca2 reversion mutations. Disruption of multiple proteins, including PARP1 and CHD4, leads to the same end point of replication fork protection, highlighting the complexities by which tumour cells evade chemotherapeutic interventions and acquire drug resistance.


Subject(s)
DNA Replication/physiology , Drug Resistance, Neoplasm/drug effects , Gene Deletion , Genes, BRCA1 , Genes, BRCA2 , Neoplasms/pathology , Nuclear Proteins/deficiency , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , DNA/biosynthesis , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Damage/drug effects , DNA Damage/genetics , DNA Helicases/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Homologous Recombination , MRE11 Homologue Protein , Mice , Neoplasms/genetics , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
11.
Front Microbiol ; 7: 688, 2016.
Article in English | MEDLINE | ID: mdl-27242714

ABSTRACT

Chronic lung infections by the ubiquitous and extremely adaptable opportunistic pathogen Pseudomonas aeruginosa correlate with the formation of a biofilm, where bacteria grow in association with an extracellular matrix and display a wide range of changes in gene expression and metabolism. This leads to increased resistance to physical stress and antibiotic therapies, while enhancing cell-to-cell communication. Oxygen diffusion through the complex biofilm structure generates an oxygen concentration gradient, leading to the appearance of anaerobic microenvironments. Ribonucleotide reductases (RNRs) are a family of highly sophisticated enzymes responsible for the synthesis of the deoxyribonucleotides, and they constitute the only de novo pathway for the formation of the building blocks needed for DNA synthesis and repair. P. aeruginosa is one of the few bacteria encoding all three known RNR classes (Ia, II, and III). Class Ia RNRs are oxygen dependent, class II are oxygen independent, and class III are oxygen sensitive. A tight control of RNR activity is essential for anaerobic growth and therefore for biofilm development. In this work we explored the role of the different RNR classes in biofilm formation under aerobic and anaerobic initial conditions and using static and continuous-flow biofilm models. We demonstrated the importance of class II and III RNR for proper cell division in biofilm development and maturation. We also determined that these classes are transcriptionally induced during biofilm formation and under anaerobic conditions. The molecular mechanism of their anaerobic regulation was also studied, finding that the Anr/Dnr system is responsible for class II RNR induction. These data can be integrated with previous knowledge about biofilms in a model where these structures are understood as a set of layers determined by oxygen concentration and contain cells with different RNR expression profiles, bringing us a step closer to the understanding of this complex growth pattern, essential for P. aeruginosa chronic infections.

12.
PLoS One ; 10(4): e0123571, 2015.
Article in English | MEDLINE | ID: mdl-25909779

ABSTRACT

Ribonucleotide reductases (RNRs) are a family of sophisticated enzymes responsible for the synthesis of the deoxyribonucleotides (dNTPs), the building blocks for DNA synthesis and repair. Although any living cell must contain one RNR activity to continue living, bacteria have the capacity to encode different RNR classes in the same genome, allowing them to adapt to different environments and growing conditions. Pseudomonas aeruginosa is well known for its adaptability and surprisingly encodes all three known RNR classes (Ia, II and III). There must be a complex transcriptional regulation network behind this RNR activity, dictating which RNR class will be expressed according to specific growing conditions. In this work, we aim to uncover the role of the transcriptional regulator NrdR in P. aeruginosa. We demonstrate that NrdR regulates all three RNR classes, being involved in differential control depending on whether the growth conditions are aerobic or anaerobic. Moreover, we also identify for the first time that NrdR is not only involved in controlling RNR expression but also regulates topoisomerase I (topA) transcription. Finally, to obtain the entire picture of NrdR regulon, we performed a global transcriptomic analysis comparing the transcription profile of wild-type and nrdR mutant strains. The results provide many new data about the regulatory network that controls P. aeruginosa RNR transcription, bringing us a step closer to the understanding of this complex system.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Gene Expression , Gene Expression Profiling , Gene Order , Genes, Reporter , Molecular Sequence Data , Mutation , Operon , Protein Binding , Pseudomonas Infections/microbiology , Ribonucleotide Reductases/genetics , Transcriptome
13.
J Control Release ; 209: 150-8, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25913364

ABSTRACT

Infections caused by biofilm-forming bacteria are a major threat to hospitalized patients and the main cause of chronic obstructive pulmonary disease and cystic fibrosis. There is an urgent necessity for novel therapeutic approaches, since current antibiotic delivery fails to eliminate biofilm-protected bacteria. In this study, ciprofloxacin-loaded poly(lactic-co-glycolic acid) nanoparticles, which were functionalized with DNase I, were fabricated using a green-solvent based method and their antibiofilm activity was assessed against Pseudomonas aeruginosa biofilms. Such nanoparticles constitute a paradigm shift in biofilm treatment, since, besides releasing ciprofloxacin in a controlled fashion, they are able to target and disassemble the biofilm by degrading the extracellular DNA that stabilize the biofilm matrix. These carriers were compared with free-soluble ciprofloxacin, and ciprofloxacin encapsulated in untreated and poly(lysine)-coated nanoparticles. DNase I-activated nanoparticles were not only able to prevent biofilm formation from planktonic bacteria, but they also successfully reduced established biofilm mass, size and living cell density, as observed in a dynamic environment in a flow cell biofilm assay. Moreover, repeated administration over three days of DNase I-coated nanoparticles encapsulating ciprofloxacin was able to reduce by 95% and then eradicate more than 99.8% of established biofilm, outperforming all the other nanoparticle formulations and the free-drug tested in this study. These promising results, together with minimal cytotoxicity as tested on J774 macrophages, allow obtaining novel antimicrobial nanoparticles, as well as provide clues to design the next generation of drug delivery devices to treat persistent bacterial infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Ciprofloxacin/administration & dosage , Deoxyribonuclease I/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Biofilms , Cell Line , Ciprofloxacin/chemistry , Ciprofloxacin/pharmacology , DNA/chemistry , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Liberation , Extracellular Matrix/drug effects , Lactic Acid/chemistry , Mice , Microbial Sensitivity Tests , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Polylysine/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
14.
Clin Cancer Res ; 20(13): 3458-71, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24799524

ABSTRACT

PURPOSE: To uncover the signaling pathways underlying follicular lymphoma-follicular dendritic cells (FL-FDC) cross-talk and its validation as new targets for therapy. EXPERIMENTAL DESIGN: FL primary cells and cell lines were cocultured in the presence or absence of FDC. After 24 and 48 hours, RNA was isolated from FL cells and subjected to gene expression profiling (GEP) and data meta-analysis using DAVID and GSEA softwares. Blockade of PI3K pathway by the pan-PI3K inhibitor BKM120 (buparlisib; Novartis Pharmaceutical Corporation) and the effect of PI3K inhibition on FL-FDC cross-talk were analyzed by means of ELISA, RT-PCR, human umbilical vein endothelial cell tube formation, adhesion and migration assays, Western blot, and in vivo studies in mouse FL xenografts. RESULTS: GEP of FL-FDC cocultures yields a marked modulation of FL transcriptome by FDC. Pathway assignment by DAVID and GSEA software uncovered an overrepresentation of genes related to angiogenesis, cell adhesion, migration, and serum-response factors. We demonstrate that the addition of the pan-PI3K inhibitor BKM120 to the cocultures was able to downregulate the expression and secretion of proangiogenic factors derived from FL-FDC cocultures, reducing in vitro and in vivo angiogenesis. Moreover, BKM120 efficiently counteracts FDC-mediated cell adhesion and impedes signaling and migration induced by the chemokine CXCL12. BKM120 inhibits both constitutive PI3K/AKT pathway and FDC- or CXCL12-induced PI3K/AKT pathway, hampers FDC survival signaling, and reduces cell proliferation of FL cells in vitro and in mouse xenografts. CONCLUSIONS: These data support the use of BKM120 in FL therapy to counteract microenvironment-related survival signaling in FL cells.


Subject(s)
Aminopyridines/pharmacology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Lymphoma, Follicular/immunology , Lymphoma, Follicular/metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Animals , Cell Adhesion/immunology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor , Cell Movement/immunology , Cell Survival/drug effects , Cell Survival/immunology , Chemokine CXCL12/metabolism , Cluster Analysis , Disease Models, Animal , Extracellular Matrix/metabolism , Gene Expression Profiling , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Internet resource in Portuguese | LIS -Health Information Locator | ID: lis-22682

ABSTRACT

Documento nº 1397 da série 'Tema para Discussão' do IPEA. O trabalho tem como objetivo documentar a relação entre renda e saúde das crianças no Brasil, usando três diferentes bases de dados e várias medidas de saúde. Na análise empírica, são utilizadas informações da Pesquisa de Orçamentos Familiares (POF) de 2002/2003, do suplemento de saúde da Pesquisa Nacional por Amostra de Domicílios (Pnad) de 2003 e da Pesquisa Nacional de Demografia e Saúde (PNDS) de 2006.


Subject(s)
Public Health , Child Welfare , Child Health , 52890 , Birth Weight , Family , Population Education , Educational Status , Income , Economic Indexes , Social Indicators , Health Status Indicators , Social Conditions , Socioeconomic Factors , Health Status , Health Evaluation , Impacts of Polution on Health , Housing , Poverty , Child , Child, Preschool
16.
Brasília; IPEA; 2009. 22 p. tab, graf.(IPEA - Texto para discussão, n. 1397).
Monography in Portuguese | LILACS | ID: lil-533679
17.
Rio de Janeiro; IPEA; 2009. 21 p. graf.(Texto para Discussão / IPEA).
Monography in Portuguese | LILACS, ECOS | ID: biblio-992010

ABSTRACT

Documenta a relação entre renda e saúde das crianças no Brasil, usando três diferentes bases de dados e várias medidas de saúde. Mostra que as crianças mais pobres tendem a ter condições de saúde piores do que as crianças mais ricas. Como crianças menos saudáveis podem ter sua capacidade produtiva reduzida no futuro, os resultados sugerem que as condições de saúde infantil no Brasil podem se constituirem importante mecanismo de transmissão intergeracional de desigualdade socioeconômica.


Subject(s)
Child , Health Statistics , Socioeconomic Factors , Health Status , Income , Maternal and Child Health , Child Health , Brazil
SELECTION OF CITATIONS
SEARCH DETAIL
...