Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Immunol ; 189(11): 5457-66, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23105143

ABSTRACT

Tumor-associated macrophages (TAMs) have been shown to promote tumor progression, and increased TAM infiltration often correlates with poor prognosis. However, questions remain regarding the phenotype of macrophages within the tumor and their role in mAb-dependent cytotoxicity. This study demonstrates that whereas TAMs have protumor properties, they maintain Fc-dependent anti-tumor function. CD11b(+)CD14(+) TAMs isolated from primary human breast tumors expressed activating FcγRs. To model breast cancer TAMs in vitro, conditioned medium from breast cancer cells was used to drive human peripheral monocyte differentiation into macrophages. Tumor-conditioned macrophages were compared with in vitro derived M1 and M2a macrophages and were found to promote tumor cell invasion and express M2a markers, confirming their protumor potential. However, unlike M2a macrophages, tumor-conditioned macrophages expressed FcγRs and phagocytosed tumor cells in the presence of a tumor Ag-targeting mAb, unmasking an underappreciated tumoricidal capacity of TAMs. In vivo macrophage depletion reduced the efficacy of anti-CD142 against MDA-MB-231 xenograft growth and metastasis in SCID/beige mice, implicating a critical role for macrophages in Fc-dependent cell killing. M-CSF was identified in tumor-conditioned media and shown to be capable of differentiating macrophages with both pro- and anti-tumor properties. These results highlight the plasticity of TAMs, which are capable of promoting tumor progression and invasion while still retaining tumoricidal function in the presence of tumor-targeting mAbs.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Macrophages/immunology , Phagocytosis , Receptors, IgG/immunology , Animals , Breast Neoplasms/pathology , CD11b Antigen/immunology , Cell Movement/drug effects , Cell Proliferation , Culture Media, Conditioned/pharmacology , Disease Progression , Female , Humans , Immunophenotyping , Lipopolysaccharide Receptors/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, SCID , Neoplasm Invasiveness/immunology , Neoplasm Transplantation , Primary Cell Culture
2.
Am J Perinatol ; 29(10): 801-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22773287

ABSTRACT

OBJECTIVE: To determine the incidence of retinopathy of prematurity (ROP) in an intensive care nursery in the Dominican Republic and to identify factors that impact ROP outcomes, screening, and treatment. STUDY DESIGN: A database analysis was performed. The database was prospectively created by the pediatric ophthalmologist in a public maternity hospital in Santo Domingo during 2009. From January to December, all infants (n = 234) who received at least one ophthalmologic examination for ROP were included. ROP screening criteria were based upon: (1) American Academy of Pediatrics guidelines and (2) the presence of critical illness in larger, more mature infants at the discretion of the neonatologist. RESULTS: Overall, 22% were diagnosed with ROP and 4.3% had severe disease. Infants with ROP had a mean birth weight of 1452 g and a mean gestational age of 31 weeks, with 35% having a gestational age >32 weeks. In multivariable regression, only gestational age remained significant (0.8, 0.68 to 0.95). Twenty-two percent diagnosed with ROP did not complete all screening procedures. CONCLUSION: There are many challenges to preventing and treating ROP in the Dominican Republic. Increased awareness of the detrimental effects of hyperoxia, broader screening criteria, and an improved screening program will help to reduce visual impairment from ROP.


Subject(s)
Intensive Care, Neonatal/methods , Neonatal Screening , Retinopathy of Prematurity , Birth Weight , Dominican Republic/epidemiology , Female , Fetal Organ Maturity , Gestational Age , Humans , Incidence , Infant, Newborn , Infant, Premature , Intensive Care Units, Neonatal/statistics & numerical data , Male , Neonatal Screening/methods , Neonatal Screening/organization & administration , Regression Analysis , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/epidemiology , Retinopathy of Prematurity/physiopathology , Risk Factors , Severity of Illness Index
3.
Cancer Epidemiol Biomarkers Prev ; 17(8): 2180-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18708412

ABSTRACT

Clinically relevant animal models of human cancer are necessary for the evaluation of putative therapeutics. We hypothesized that circulating human lung cancer-associated proteins would correlate with physiologic measurements from an orthotopic H460 human non-small cell lung carcinoma model that we developed in immunodeficient rats. Physiologic measurements and serum samples were collected over time. Serum interleukin-8 (IL-8), p53, vascular endothelial growth factor, and matrix metalloproteinase-9 were quantitated for correlation with physiologic measurements. Matrix metalloproteinase-9 and p53 were not significantly detectable. Circulating vascular endothelial growth factor was detected at high levels in some tumor-bearing animals. Human IL-8 was detectable in all tumor-bearing animals and correlated positively with markers of respiratory acidosis (pH, P = 0.012; TCO(2), P = 0.024; pCO(2), P = 0.007; and HCO(3)(-), P = 0.029) and with surface body temperature (P = 0.001) beginning on day 16 after implantation. IL-8 levels negatively correlated with survival (P < 0.001), indicating an association with tumor burden. Circulating human IL-8 might be a useful, clinically relevant circulating tumor protein marker due to its positive correlation with multiple physiologic variables associated with lung cancer progression.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/blood , Interleukin-8/blood , Lung Neoplasms/blood , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Matrix Metalloproteinase 9/blood , Proportional Hazards Models , Rats , Rats, Nude , Tumor Cells, Cultured , Tumor Suppressor Protein p53/blood , Vascular Endothelial Growth Factor A/blood
4.
Clin Exp Metastasis ; 25(2): 139-48, 2008.
Article in English | MEDLINE | ID: mdl-18064530

ABSTRACT

CNTO 95 is a fully human monoclonal antibody that recognizes alphav integrins. Previous studies have shown that CNTO 95 exhibits both anti-tumor and anti-angiogenic activities (Trikha M et al., Int J Cancer 110:326-335, 2004). In this study we investigated the biological activities of CNTO 95 on breast tumor cells both in vitro and in vivo. In vitro treatment with CNTO 95 decreased the viability of breast tumor cells adhering to vitronectin. CNTO 95 inhibited tumor cell adhesion, migration, and invasion in vitro. CNTO 95 treatment also induced tyrosine dephosphorylation of focal adhesion kinase (FAK), and the docking protein paxillin that recruits both structural and signaling molecules to focal adhesions (Turner CE, Int J Biochem Cell Biol 30:955-959, 1998; O'Neil GM et al., Trends Cell Biol 10:111-119, 2000). These results suggest that CNTO 95 inhibits breast tumor cell growth, migration and invasion by interruption of alphav integrin mediated focal adhesions and cell motility signals. In vivo studies of CNTO 95 were conducted in an orthotopic breast tumor xenograft model. Treatment with CNTO 95 resulted in significant inhibition of both tumor growth and spontaneous metastasis of MDA-MB-231 cells to the lungs. CNTO 95 also inhibited lung metastasis in a separate experimental (tail vein injection) model of metastasis. The results presented here demonstrate the anti-tumor and anti-metastatic activities of CNTO 95 in breast cancer models and provide insight into the cellular and molecular mechanisms mediating its inhibitory effects on metastasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/metabolism , Cell Movement/drug effects , Integrin alphaV/immunology , Animals , Antibodies, Monoclonal, Humanized , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Female , Focal Adhesion Kinase 1/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, SCID , Neoplasm Invasiveness , Paxillin/metabolism , Phosphorylation , Signal Transduction/drug effects , Transplantation, Heterologous , Vitronectin/metabolism
5.
Clin Cancer Res ; 13(12): 3689-95, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575234

ABSTRACT

PURPOSE: Targeted delivery of cytotoxic agents to solid tumors through cell surface antigens can potentially reduce systemic toxicity and increase the efficacy of the targeted compounds. The purpose of this study was to show the feasibility of treating solid tumors by targeting alpha(v) integrins with antibody-maytansinoid conjugates and to test the relative in vivo activities of several linker-maytansinoid chemistries. EXPERIMENTAL DESIGN: CNTO 364, CNTO 365, and CNTO 366 are targeted cytotoxic agents created by conjugating the CNTO 95 anti-alpha(v) integrin antibody with three distinct maytansinoid-linker structures. These structures were designed to have varying degrees of chemical substitution surrounding the disulfide bond linking the cytotoxic agent to the antibody. A model conjugate was shown to be specifically cytotoxic in vitro and highly active against established human tumor xenografts in immunocompromised rats. The in vivo antitumor activities of CNTO 364, CNTO 365, and CNTO 366 were compared in rat xenograft models. RESULTS: CNTO 365, with a linker chemistry of expected intermediate stability, was shown to be substantially more active than the other two conjugates with lesser or greater substitution around the disulfide linkage. CONCLUSION: CNTO 95-maytansinoid immunoconjugates are potent antitumor agents against alpha(v) integrin-expressing human carcinomas. These studies show for the first time the feasibility of targeting alpha(v) integrins on solid tumors with tumor-activated prodrugs. The DM4 linker-maytansinoid configuration of CNTO 365 was substantially more active in the models tested here when compared with alternative configurations with greater or lesser chemical substitution surrounding the linker.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Immunoconjugates/administration & dosage , Integrin alpha5/immunology , Neoplasms, Experimental/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Delivery Systems , Female , Flow Cytometry , Humans , Immunoconjugates/chemistry , Immunotherapy , Mice , Rats , Xenograft Model Antitumor Assays
6.
Int J Cancer ; 120(6): 1261-7, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17192924

ABSTRACT

Thromboembolic complications are frequently associated with advanced cancer. Interestingly, one of the major initiators of blood coagulation, tissue factor (TF), is reported to be overexpressed in several tumor types and can be found on both tumor cells and tumor vasculature. Although the exact mechanisms have yet to be elucidated, TF expressed on tumor cells can trigger intracellular signaling events through various pathways that can lead to tumor angiogenesis, proliferation, and metastasis. There exists preclinical evidence that disruption of TF dependent signaling can effectively inhibit tumor cell migration, metastasis, and angiogenesis. Here, we report for the first time that an antibody to tissue factor can also prevent tumor growth in vivo. Prophylactic administration of CNTO 859, a humanized anti-human TF antibody, was shown to inhibit experimental lung metastasis of MDA-MB-231 human breast carcinoma cells by over 99% compared to a control antibody. Furthermore, therapeutic doses of CNTO 859 were shown to reduce tumor incidence and growth of orthotopically implanted MDA-MB-231 cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Immunoglobulin G/therapeutic use , Lung Neoplasms/drug therapy , Thromboplastin/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , Carcinoma/prevention & control , Carcinoma/secondary , Cell Proliferation/drug effects , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred Strains , Xenograft Model Antitumor Assays
7.
Mol Cancer Res ; 4(6): 371-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16778084

ABSTRACT

Extracellular matrix metalloproteinase (MMP) inducer (EMMPRIN) is a cell surface glycoprotein overexpressed in many solid tumors. In addition to its ability to stimulate stromal MMP expression, tumor-associated EMMPRIN also induces vascular endothelial growth factor (VEGF) expression. To explore the underlying signaling pathways used by EMMPRIN, we studied the involvement of phosphoinositide 3-kinase (PI3K)-Akt, mitogen-activated protein kinase (MAPK), JUN, and p38 kinases in EMMPRIN-mediated VEGF regulation. Overexpression of EMMPRIN in MDA-MB-231 breast cancer cells stimulated the phosphorylation of only Akt and MAPKs but not that of JUN and p38 kinases. Conversely, inhibition of EMMPRIN expression resulted in suppressed Akt and MAPK phosphorylation. Furthermore, the PI3K-specific inhibitor LY294002 inhibited VEGF production by EMMPRIN-overexpressing cells in a dose- and time-dependent manner. On the other hand, the MAPK inhibitor U0126 did not affect VEGF production. In vivo, EMMPRIN-overexpressing tumors with elevated VEGF expression had a high level of phosphorylation of Akt and MAPK. Finally, when fibroblast cells were treated with recombinant EMMPRIN, Akt kinase but not MAPK was phosphorylated concomitant with an increase in VEGF production. Both the activation of Akt kinase and the induction of VEGF were specifically inhibited with a neutralizing antibody to EMMPRIN. Our results show that in both tumor and fibroblast cells EMMPRIN regulates VEGF production via the PI3K-Akt pathway but not via the MAPK, JUN, or p38 kinase pathways.


Subject(s)
Basigin/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Basigin/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Matrix Metalloproteinases , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Neovascularization, Pathologic , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Proteins/pharmacology , Transplantation, Heterologous , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Cancer Res ; 65(8): 3193-9, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833850

ABSTRACT

Matrix metalloproteinases (MMPs) are endopeptidases that play pivotal roles in promoting tumor disease progression, including tumor angiogenesis. In many solid tumors, MMP expression could be attributed to tumor stromal cells and is partially regulated by tumor-stroma interactions via tumor cell-associated extracellular matrix metalloproteinase inducer (EMMPRIN). The role of EMMPRIN during tumor angiogenesis and growth was explored by modulating EMMPRIN expression and activity using recombinant DNA engineering and neutralizing antibodies. In human breast cancer cells, changes in EMMPRIN expression influenced vascular endothelial growth factor (VEGF) production at both RNA and protein levels. In coculture of tumor cells and fibroblasts mimicking tumor-stroma interactions, VEGF expression was induced in an EMMPRIN- and MMP-dependent fashion, and was further enhanced by overexpressing EMMPRIN. Conversely, VEGF expression was inhibited by suppressing EMMPRIN expression in tumor cells, by neutralizing EMMPRIN activity, or by inhibiting MMPs. In vivo, EMMPRIN overexpression stimulated tumor angiogenesis and growth; both were significantly inhibited by antisense suppression of EMMPRIN. Expression of both human and mouse VEGF and MMP, derived from tumor and host cells, respectively, was regulated by EMMPRIN. These results suggest a novel tumor angiogenesis mechanism in which tumor-associated EMMPRIN functionally mediates tumor-stroma interactions and directly contributes to tumor angiogenesis and growth by stimulating VEGF and MMP expression.


Subject(s)
Antigens, CD/physiology , Breast Neoplasms/blood supply , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Basigin , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Coculture Techniques , Endothelial Cells/cytology , Female , Fibroblasts/cytology , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/antagonists & inhibitors
9.
J Immunother ; 25 Suppl 1: S20-7, 2002.
Article in English | MEDLINE | ID: mdl-12048347

ABSTRACT

Interleukin (IL)-18 has profound antitumor activity when administered at high doses as a single agent for prolonged periods in BALB/c mice bearing late, well-established MOPC-315 tumors. Management with a qD x 27 schedule resulted in regression of tumors in all animals receiving 5 mg/kg/d. A protracted daily management regimen appears to be necessary to induce regression in this advanced tumor model. Biologic markers were assessed and appear to be potentially useful in evaluating the immunologic and antitumor activity of IL-18. The biomarkers of IL-18's immunologic activity include, but are not limited to, IL-1alpha, IL-2, IL-8, IL-10, IL-12, IL-13, interferon-gamma, tumor necrosis factor-alpha, and granulocyte-macrophage colony-stimulating factor. The profile of these circulating cytokines and their expression levels at baseline, and after IL-18 delivery, can be measured in the serum, as well as from splenocytes of mice or human peripheral blood mononuclear cells derived from either normal subjects or patients with cancer. We compared IL-18 and IL-12 alone or in combination for their ability to induce cytokine production and natural killer cytolytic activity. Our data support the notion that IL-18 induces a predominantly Th1 response, and that the mechanism of IL-18 activity differs from that of IL-12. The biologic activity of IL-18 management revealed by increases in serum levels of cytokines and enhancement of natural killer cytolytic activity will be useful as clinical trials initiate in 2002. Expression of interferon-gamma and granulocyte-macrophage colony-stimulating factor serum levels correlates directly over a broad dose escalation with the level of IL-18. Therefore, this provides a convenient pharmacodynamic reference to the biologic response to IL-18 that may serve to guide the conduct of clinical trials.


Subject(s)
Cytokines/metabolism , Immunity, Cellular/drug effects , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Plasmacytoma/drug therapy , Th1 Cells/drug effects , Animals , Cytokines/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Recombination, Genetic , Reference Values , Sensitivity and Specificity , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...