Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
JCO Glob Oncol ; 10: e2300216, 2024 May.
Article in English | MEDLINE | ID: mdl-38723219

ABSTRACT

PURPOSE: Breast cancer mortality rates in Latin America (LA) are higher than those in the United States, possibly because of advanced disease presentation, health care disparities, or unfavorable molecular subtypes. The Latin American Cancer Research Network was established to address these challenges and to promote collaborative clinical research. The Molecular Profiling of Breast Cancer Study (MPBCS) aimed to evaluate the clinical characteristics and treatment outcomes of LA participants with locally advanced breast cancer (LABC). PATIENTS AND METHODS: The MPBCS enrolled 1,449 participants from Argentina, Brazil, Chile, Mexico, and Uruguay. Through harmonized procedures and quality assurance measures, this study evaluated clinicopathologic characteristics, neoadjuvant chemotherapy response, and survival outcomes according to residual cancer burden (RCB) and the type of surgery. RESULTS: Overall, 711 and 480 participants in the primary surgery and neoadjuvant arms, respectively, completed the 5-year follow-up period. Overall survival was independently associated with RCB (worse survival for RCBIII-adjusted hazard ratio, 8.19, P < .001, and RCBII [adjusted hazard ratio, 3.69, P < .008] compared with RCB0 [pathologic complete response or pCR]) and type of surgery (worse survival in mastectomy than in breast-conserving surgery [BCS], adjusted hazard ratio, 2.97, P = .001). The hormone receptor-negative-human epidermal growth factor receptor 2-positive group had the highest proportion of pCR (48.9%). The analysis of the ASCO Quality Oncology Practice Initiative breast module revealed high compliance with pathologic standards but lower adherence to treatment administration standards. Notably, compliance with trastuzumab administration varied widely among countries (33.3%-88.7%). CONCLUSION: In LABC, we demonstrated the survival benefit of BCS and the prognostic effect of the response to available neoadjuvant treatments despite an important variability in access to key treatments. The MPBCS represents a significant step forward in understanding the real-world implementation of oncologic procedures in LA.


Subject(s)
Breast Neoplasms , Neoadjuvant Therapy , Humans , Breast Neoplasms/therapy , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Female , Middle Aged , Latin America/epidemiology , Adult , Aged
2.
Front Oncol ; 12: 845527, 2022.
Article in English | MEDLINE | ID: mdl-35530311

ABSTRACT

Molecular profile of breast cancer in Latin-American women was studied in five countries: Argentina, Brazil, Chile, Mexico, and Uruguay. Data about socioeconomic characteristics, risk factors, prognostic factors, and molecular subtypes were described, and the 60-month overall cumulative survival probabilities (OS) were estimated. From 2011 to 2013, 1,300 eligible Latin-American women 18 years or older, with a diagnosis of breast cancer in clinical stage II or III, and performance status ≦̸1 were invited to participate in a prospective cohort study. Face-to-face interviews were conducted, and clinical and outcome data, including death, were extracted from medical records. Unadjusted associations were evaluated by Chi-squared and Fisher's exact tests and the OS by Kaplan-Meier method. Log-rank test was used to determine differences between cumulative probability curves. Multivariable adjustment was carried out by entering potential confounders in the Cox regression model. The OS at 60 months was 83.9%. Multivariable-adjusted death hazard differences were found for women living in Argentina (2.27), Chile (1.95), and Uruguay (2.42) compared with Mexican women, for older (≥60 years) (1.84) compared with younger (≤40 years) women, for basal-like subtype (5.8), luminal B (2.43), and HER2-enriched (2.52) compared with luminal A subtype, and for tumor clinical stages IIB (1.91), IIIA (3.54), and IIIB (3.94) compared with stage IIA women. OS was associated with country of residence, PAM50 intrinsic subtype, age, and tumor stage at diagnosis. While the latter is known to be influenced by access to care, including cancer screening, timely diagnosis and treatment, including access to more effective treatment protocols, it may also influence epigenetic changes that, potentially, impact molecular subtypes. Data derived from heretofore understudied populations with unique geographic ancestry and sociocultural experiences are critical to furthering our understanding of this complexity.

3.
Mol Cancer Res ; 15(3): 304-316, 2017 03.
Article in English | MEDLINE | ID: mdl-28031408

ABSTRACT

Understanding the mechanism of metastatic dissemination is crucial for the rational design of novel therapeutics. The secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein which has been extensively associated with human breast cancer aggressiveness although the underlying mechanisms are still unclear. Here, shRNA-mediated SPARC knockdown greatly reduced primary tumor growth and completely abolished lung colonization of murine 4T1 and LM3 breast malignant cells implanted in syngeneic BALB/c mice. A comprehensive study including global transcriptomic analysis followed by biological validations confirmed that SPARC induces primary tumor growth by enhancing cell cycle and by promoting a COX-2-mediated expansion of myeloid-derived suppressor cells (MDSC). The role of SPARC in metastasis involved a COX-2-independent enhancement of cell disengagement from the primary tumor and adherence to the lungs that fostered metastasis implantation. Interestingly, SPARC-driven gene expression signatures obtained from these murine models predicted the clinical outcome of patients with HER2-enriched breast cancer subtypes. In total, the results reveal that SPARC and its downstream effectors are attractive targets for antimetastatic therapies in breast cancer.Implications: These findings shed light on the prometastatic role of SPARC, a key protein expressed by breast cancer cells and surrounding stroma, with important consequences for disease outcome. Mol Cancer Res; 15(3); 304-16. ©2016 AACR.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Osteonectin/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Osteonectin/genetics , Prognosis , Receptor, ErbB-2/genetics , Treatment Outcome
4.
Biochim Biophys Acta ; 1860(10): 2255-68, 2016 10.
Article in English | MEDLINE | ID: mdl-27130882

ABSTRACT

BACKGROUND: We previously demonstrated that the activated leukocyte cell adhesion molecule (ALCAM/CD166) can interact with galectin-8 (Gal-8) in endothelial cells. ALCAM is a member of the immunoglobulin superfamily that promotes homophilic and heterophilic cell-cell interactions. Gal-8 is a "tandem-repeat"-type galectin, known as a matricellular protein involved in cell adhesion. Here, we analyzed the physical interaction between both molecules in breast cancer cells and the functional relevance of this phenomenon. METHODS: We performed binding assays by surface plasmon resonance to study the interaction between Gal-8 and the recombinant glycosylated ALCAM ectodomain or endogenous ALCAM from MDA-MB-231 breast cancer cells. We also analyzed the binding of ALCAM-silenced or control breast cancer cells to immobilized Gal-8 by SPR. In internalization assays, we evaluated the influence of Gal-8 on ALCAM surface localization. RESULTS: We showed that recombinant glycosylated ALCAM and endogenous ALCAM from breast carcinoma cells physically interacted with Gal-8 in a glycosylation-dependent fashion displaying a differential behavior compared to non-glycosylated ALCAM. Moreover, ALCAM-silenced breast cancer cells exhibited reduced binding to Gal-8 relative to control cells. Importantly, exogenously added Gal-8 provoked ALCAM segregation, probably trapping this adhesion molecule at the surface of breast cancer cells. CONCLUSIONS: Our data indicate that Gal-8 interacts with ALCAM at the surface of breast cancer cells through glycosylation-dependent mechanisms. GENERAL SIGNIFICANCE: A novel heterophilic interaction between ALCAM and Gal-8 is demonstrated here, suggesting its physiologic relevance in the biology of breast cancer cells.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Galectins/metabolism , Protein Interaction Maps/genetics , Antigens, CD/genetics , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication/genetics , Cell Line, Tumor , Cell Movement/genetics , Endothelial Cells/metabolism , Female , Fetal Proteins/genetics , Galectins/genetics , Glycosylation , Humans , Protein Binding , Surface Properties
5.
Eur J Immunol ; 45(5): 1560-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25726929

ABSTRACT

Clinical studies suggest that triple negative breast cancer (TNBC) patients with epidermal growth factor receptor (EGFR)-expressing tumors could benefit from therapy with Cetuximab, which targets EGFR. NK cells are the primary effectors of antibody (Ab)-dependent cell-mediated cytotoxicity (ADCC) and thus play a role in Ab-based therapies. We have previously described diminished levels of Cetuximab-mediated ADCC in vitro in patients with advanced breast cancer. Here, we investigated the potential causes of this NK-cell functional deficiency. We characterized NK-cell activating/inhibitory receptors in the peripheral blood of breast cancer patients and found CD85j inhibitory receptor overexpression. The capacity of NK cells to perform Cetuximab-triggered ADCC against TNBC cells correlated inversely with CD85j expression, even in the presence of the stimulatory cytokines IL-2 or IL-15. Hence, patients expressing high levels of CD85j had an impaired ability to lyse TNBC cells in the presence of Cetuximab. We also found that CD85j overexpression was associated with HLA-I and soluble HLA-G expression by tumors. A CD85j functional blockade with a CD85j antagonist Ab restored ADCC levels in breast cancer patients and reverted this negative effect. Our data suggest that strategies that overcome the hurdles of immune activation could improve Cetuximab clinical efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD/metabolism , Killer Cells, Natural/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Adult , Antineoplastic Agents/pharmacology , Case-Control Studies , Cetuximab , ErbB Receptors/antagonists & inhibitors , Female , HLA Antigens/metabolism , HLA-G Antigens/metabolism , Humans , K562 Cells , Leukocyte Immunoglobulin-like Receptor B1 , Middle Aged , Young Adult
6.
Int J Exp Pathol ; 95(3): 181-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24602133

ABSTRACT

Proteoglycan accumulation within the arterial intima has been implicated in atherosclerosis progression in humans. Nevertheless, hypercholesterolaemia is unable to induce intimal thickening and atheroma plaque development in rats. The study was performed to analyse proteoglycans modifications in rats fed with a high-cholesterol diet to understand whether vascular wall remodelling protects against lesions. Sections obtained from rat aortas showed normal features, in intimal-to-media ratio and lipid accumulation. However, focal endothelial hyperplasia and neo-intima rearrangement were observed in high-cholesterol animals. Besides, hypercholesterolaemia induced an inflammatory microenviroment. We determined the expression of different proteoglycans from aortic cells by Western blot and observed a diminished production of decorin and biglycan in high-cholesterol animals compared with control (P < 0.01 and P < 0.05, respectively). Versican was increased in high-cholesterol animals (P < 0.05), whereas perlecan production showed no differences. No modification of the total content of glycosaminoglycans (GAGs) was found between the two experimental groups. In contrast, the chondroitin sulphate/dermatan sulphate ratio was increased in the high-cholesterol group as compared to the control (0.56 and 0.34, respectively). Structural alterations in the disaccharide composition of galactosaminoglycans were also detected by HPLC, as the ratio of 6-sulphate to 4-sulphate disaccharides was increased in high-cholesterol animals (P < 0.05). Our results suggest that attenuation of decorin and biglycan expression might be an effective strategy to inhibit the first step in atherogenesis, although specific GAG structural modification associated with the development of vascular disease took place. Results emphasize the potential application of therapies based on vascular matrix remodelling to treat atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Chondroitin Sulfate Proteoglycans/metabolism , Dermatan Sulfate/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Hypercholesterolemia/physiopathology , Plaque, Atherosclerotic/prevention & control , Animals , Aorta/cytology , Aorta/metabolism , Atherosclerosis/physiopathology , Cholesterol/blood , Chondroitin Sulfate Proteoglycans/chemistry , Dermatan Sulfate/chemistry , Diet, Atherogenic/adverse effects , Disease Models, Animal , Glycosaminoglycans/chemistry , Goats , Humans , Hypercholesterolemia/metabolism , Lipids/blood , Male , Rabbits , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Breast Cancer Res Treat ; 136(3): 659-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065032

ABSTRACT

Triple-negative breast cancer (TNBC) patients do not benefit from target-specific treatments and is associated with a high relapse rate. Epidermal growth factor receptor is frequently expressed in TNBC and is a candidate for new therapies. In this work, we studied Cetuximab-mediated immune activity by NK cells. Thirteen activating/inhibitory receptors were examined on peripheral blood and tumor infiltrating NK cells. NK-cell functionality was evaluated using as effectors tumor-modulated NK cells and NK cells from patients. We evaluated the treatment with Cetuximab plus IL-2 or IL-15 in vivo in TNBC xenografts. Tumor NK-cells receptor profile showed upregulation of inhibitory receptors and downregulation of activating ones. Tumor-modulated NK cells were less cytotoxic. They could perform antibody-dependent cellular cytotoxicity (ADCC) triggered by Cetuximab, although impaired, it could still be restored by stimulation with IL-2 or IL-15. Patients with advanced disease displayed diminished levels of ADCC compared to healthy volunteers. ADCC was restored and potentiated with both cytokines, which were also effective in enhancing the therapeutic activity of Cetuximab in vivo. The combination of Cetuximab with IL-15 and IL-2 may be considered an attractive therapeutic approach to enhance the clinical efficacy of Cetuximab in TNBC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Adult , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cetuximab , Female , Humans , Mice , Mice, Nude , Middle Aged , Xenograft Model Antitumor Assays
8.
Mol Ther ; 20(12): 2222-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22948673

ABSTRACT

Targeting the tumor stroma in addition to the malignant cell compartment is of paramount importance to achieve complete tumor regression. In this work, we modified a previously designed tumor stroma-targeted conditionally replicative adenovirus (CRAd) based on the SPARC promoter by introducing a mutated E1A unable to bind pRB and pseudotyped with a chimeric Ad5/3 fiber (Ad F512v1), and assessed its replication/lytic capacity in ovary cancer in vitro and in vivo. AdF512v1 was able to replicate in fresh samples obtained from patients: (i) with primary human ovary cancer; (ii) that underwent neoadjuvant treatment; (iii) with metastatic disease. In addition, we show that four intraperitoneal (i.p.) injections of 5 × 10(10) v.p. eliminated 50% of xenografted human ovary tumors disseminated in nude mice. Moreover, AdF512v1 replication in tumor models was enhanced 15-40-fold when the tumor contained a mix of malignant and SPARC-expressing stromal cells (fibroblasts and endothelial cells). Contrary to the wild-type virus, AdF512v1 was unable to replicate in normal human ovary samples while the wild-type virus can replicate. This study provides evidence on the lytic capacity of this CRAd and highlights the importance of targeting the stromal tissue in addition to the malignant cell compartment to achieve tumor regression.


Subject(s)
Adenovirus E1A Proteins/genetics , Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Humans , Mice , Ovarian Neoplasms/genetics , Stromal Cells/metabolism , Xenograft Model Antitumor Assays
9.
J Invest Dermatol ; 132(2): 365-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21993558

ABSTRACT

MART-1 and gp100 are prototypical melanoma antigen (Ag), but their clinical use as vaccines or as targets of cytotoxic lymphocytes achieved modest success. Possible explanations could be that as MART-1 and gp100 are melanocyte differentiation Ag, clonogenic Ag-non-expressing cells would be spared by immune effectors, or that clonogenic cells would be intrinsically resistant to cytotoxic lymphocytes. We therefore analyzed the proliferative status of MART-1/gp100-expressing and -non-expressing cells in biopsies, and the clonogenicity and sensitiveness to cytotoxic lymphocytes of the human cutaneous melanoma cell lines MEL-XY1 and MEL-XY3. Analysis of MART-1/gp100 and Ki-67 expression in 22 melanoma tumors revealed that MART-1/gp100-expressing and -non-expressing cells proliferated competitively. MART-1, gp100, tyrosinase, and CD271 expression were studied in MEL-XY1 and MEL-XY3 colonies. At 7 days, colonies displayed positive, negative, and mixed expression patterns. By 14 days, colonies of different sizes developed, showing cells with different clonogenic potential, and Ag were downregulated, suggesting Ag plasticity. Subcloning of MEL-XY1 colonies showed that Ag expression varied with time without interfering with clonogenicity. Finally, clonogenic, MART-1/gp100-expressing cells were lysed by specific CD8 lymphocytes. Thus, MART-1 and gp100 expression and plasticity would not interfere with proliferation or clonogenicity, and clonogenic cells may be lysed by cytotoxic lymphocytes.


Subject(s)
Cell Proliferation , MART-1 Antigen/analysis , Melanoma/pathology , Skin Neoplasms/pathology , gp100 Melanoma Antigen/analysis , DNA Methylation , Humans , Ki-67 Antigen/analysis , MART-1 Antigen/genetics , MART-1 Antigen/physiology , Melanoma/chemistry , Promoter Regions, Genetic , Skin Neoplasms/chemistry , T-Lymphocytes, Cytotoxic/immunology , gp100 Melanoma Antigen/physiology
10.
Vaccine ; 28(51): 8162-8, 2010 Nov 29.
Article in English | MEDLINE | ID: mdl-20937314

ABSTRACT

Antigen presentation by dendritic cells (DC) is of key importance for the initiation of the primary immune response. Mice vaccinated with DC charged with apoptotic/necrotic B16 cells (DC-Apo/Nec) are protected against B16 challenge. The aim of this study was to assess vaccine cell migration in our system and to find out if there is an immunological response taking place at the vaccination site. The formation of a pseudocapsule, peripheral node addresin expression in small venules, and the recruitment of a wide variety of cellular populations, including macrophages, polymorphonuclear lymphocytes, and CD8+ and CD4+ T lymphocytes found in association with DC, evidenced the formation of tertiary lymphoid tissue in the vaccination site in our experimental system.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Lymphoid Tissue/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Vaccination/methods , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Histocytochemistry , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Subcutaneous Tissue/immunology , Survival Analysis
11.
PLoS One ; 4(4): e5119, 2009.
Article in English | MEDLINE | ID: mdl-19337591

ABSTRACT

The clinical efficacy of conditionally replicative oncolytic adenoviruses (CRAd) is still limited by the inefficient infection of the tumor mass. Since tumor growth is essentially the result of a continuous cross-talk between malignant and tumor-associated stromal cells, targeting both cell compartments may profoundly influence viral efficacy. Therefore, we developed SPARC promoter-based CRAds since the SPARC gene is expressed both in malignant cells and in tumor-associated stromal cells. These CRAds, expressing or not the Herpes Simplex thymidine kinase gene (Ad-F512 and Ad(I)-F512-TK, respectively) exerted a lytic effect on a panel of human melanoma cells expressing SPARC; but they were completely attenuated in normal cells of different origins, including fresh melanocytes, regardless of whether cells expressed or not SPARC. Interestingly, both CRAds displayed cytotoxic activity on SPARC positive-transformed human microendothelial HMEC-1 cells and WI-38 fetal fibroblasts. Both CRAds were therapeutically effective on SPARC positive-human melanoma tumors growing in nude mice but exhibited restricted efficacy in the presence of co-administered HMEC-1 or WI-38 cells. Conversely, co-administration of HMEC-1 cells enhanced the oncolytic efficacy of Ad(I)-F512-TK on SPARC-negative MIA PaCa-2 pancreatic cancer cells in vivo. Moreover, conditioned media produced by stromal cells pre-infected with the CRAds enhanced the in vitro viral oncolytic activity on pancreatic cancer cells, but not on melanoma cells. The whole data indicate that stromal cells might play an important role on the outcome of the oncolytic efficacy of conditionally replicative adenoviruses.


Subject(s)
Adenoviridae/physiology , Neoplasms/pathology , Oncolytic Virotherapy , Stromal Cells/pathology , Animals , Cell Line , Cell Line, Tumor , Cytopathogenic Effect, Viral , Humans , Mice , Neoplasm Transplantation , Neoplasms/therapy , Osteonectin/genetics , Promoter Regions, Genetic , Virus Replication
12.
J Invest Dermatol ; 127(11): 2618-28, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17625595

ABSTRACT

SPARC (secreted protein acidic and rich in cysteine) is a matricellular protein whose overexpression in malignant or tumor-stromal cells is often associated with increased aggressiveness and bad prognosis in a wide range of human cancer types, particularly melanoma. We established the impact that changes in the level of SPARC produced by malignant cells and neighboring stromal cells have on melanoma growth. Melanoma cell growth in monolayer was only slightly affected by changes in SPARC levels. However, melanoma growth in spheroids was strongly inhibited upon SPARC hyperexpression and conversely enhanced when SPARC expression was downregulated. Interestingly, SPARC overexpression in neighboring fibroblasts had no effect on spheroid growth irrespective of SPARC levels expressed by the melanoma cells, themselves. Downregulation of SPARC expression in melanoma cells induced their rejection in vivo through a mechanism mediated exclusively by host polymorphonuclear cells. On the other hand, SPARC hyperexpression enhanced vascular density, collagen deposition, and fibroblast recruitment in the surrounding stroma without affecting melanoma growth. In agreement with the in vitro data, overexpression of SPARC in co-injected fibroblasts did not affect melanoma growth in vivo. All the data indicate that melanoma growth is not subject to regulation by exogenous SPARC, nor by stromal organization, but only by SPARC levels produced by the malignant cells themselves.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Osteonectin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Adenoviridae/genetics , Cell Line, Tumor , Cell Proliferation , Collagen/metabolism , Disease Progression , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Osteonectin/genetics , Skin Neoplasms/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Transduction, Genetic
13.
J Invest Dermatol ; 127(8): 2031-41, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17460736

ABSTRACT

Chemokines such as monocyte chemoattractant protein (MCP)-1 are key agonists that attract macrophages to tumors. In melanoma, it has been previously shown that variable levels of MCP-1/CCL2 appear to correlate with infiltrating macrophages and tumor fate, with low to intermediate levels of the chemokine contributing to melanoma development. To work under such conditions, a poorly tumorigenic human melanoma cell line was transfected with an expression vector encoding MCP-1. We found that M2 macrophages are associated to MCP-1+ tumors, triggering a profuse vascular network. To target the protumoral macrophages recruitment and reverting tumor growth promotion, clodronate-laden liposomes (Clod-Lip) or bindarit were administered to melanoma-bearing mice. Macrophage depletion after Clod-Lip treatment induced development of smaller tumors than in untreated mice. Immunohistochemical analysis with an anti-CD31 antibody revealed scarce vascular structures mainly characterized by narrow vascular lights. Pharmacological inhibition of MCP-1 with bindarit also reduced tumor growth and macrophage recruitment, rendering necrotic tumor masses. We suggest that bindarit or Clod-Lip abrogates protumoral-associated macrophages in human melanoma xenografts and could be considered as complementary approaches to antiangiogenic therapy.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Clodronic Acid/administration & dosage , Indazoles/therapeutic use , Macrophages/physiology , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Propionates/therapeutic use , Animals , Cell Line, Tumor , Chemokine CCL2/physiology , Humans , Liposomes , Male , Melanoma, Experimental/blood supply , Mice , Neoplasm Transplantation , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Transplantation, Heterologous
14.
J Immunother ; 29(4): 444-54, 2006.
Article in English | MEDLINE | ID: mdl-16799340

ABSTRACT

We investigated whether recombinant human granulocyte-monocyte-colony-stimulating factor (rhGM-CSF) increased the immunogenicity of VACCIMEL, a vaccine consisting of 3 irradiated allogeneic melanoma cell lines. A phase I clinical trial was performed on 20 melanoma patients in stages IIB (n=2), III (n=10), and IV (n=8), who were disease free after surgery (n=16) or had minimal disease (n=4). Cohorts of 4 patients were vaccinated 4 times with VACCIMEL and bacillus Calmette Guerin (BCG) as adjuvant. Besides, the patients received placebo (group 1) or GM-CSF: 150 microg (group 2), 300 microg (group 3), 400 microg (group 4), and 600 microg (group 5) per vaccine. The combination of VACCIMEL and GM-CSF had low toxicity. Only in group 5, grade 2 thoracic pain (3/4 patients) and abdominal cramps (2/4 patients) were observed. Delayed-type hypersensitivity increased after vaccination and it was highest in group 4. Phytohemagglutinin stimulation of peripheral blood lymphocytes was analyzed in 9 patients: 4/9 had normal stimulation; 3/9 had low basal stimulation, which recovered after vaccination; and 2/9 were not stimulated. Antimelanoma antibodies preexisted in 9/19 patients; in 3/19 patients, antibodies anti-33 kd, 90 kd, and 100 kd antigens were induced by vaccination. IgG2 but not IgG1 antibodies were detected. Anti-BCG antibodies, mostly IgG2, reached the highest post/prevaccination ratio in group 4. Median serum interleukin-12 was lower in progressing patients (61.6 pg/mL) than in those without evident disease (89 pg/mL). Thus, its low toxicity and the induction of a predominantly cellular immune response suggest that the addition of 300 to 400 microg GM-CSF to VACCIMEL is useful in increasing the immune response.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Melanoma/drug therapy , Melanoma/prevention & control , Abdominal Pain/chemically induced , Administration, Cutaneous , Adolescent , Adult , Aged , Antibodies, Neoplasm/immunology , BCG Vaccine/adverse effects , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Fatigue/chemically induced , Female , Humans , Immunotherapy, Adoptive/methods , Interleukin-10/blood , Interleukin-12/blood , Lymphocyte Activation/drug effects , Lymphocyte Activation/physiology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma/pathology , Middle Aged , Phytohemagglutinins/pharmacology , Recombinant Proteins
15.
Rev. argent. cardiol ; 74(2): 149-156, mar.-abr. 2006. graf
Article in Spanish | LILACS | ID: lil-436478

ABSTRACT

Las comunicaciones intercelulares son eventos biológicos esenciales en los organismos multicelulares, asociadas con el control del crecimiento y la diferenciación celular, la apoptosis, las respuestas adaptativas de células diferenciadas y la sincronización de funciones celulares. Las uniones intercelulares, conocidas como uniones gap, estructuralmente constituidas por conexinas, tienen una participación activa en estos procesos. A nivel cardiovascular, la comunicación célula a célula es indispensable, en condiciones normales, para la embriogénesis cardíaca, la transmisión del impulso eléctrico, la sincronización de la actividad contráctil cardíaca, la transmisión de señales reflejas vasculares, entre otras funciones biológicas, mientras que en condiciones patológicas, a causa de mutaciones genéticas heredadas o adquiridas, participan en el desarrollo de cardiopatías congénitas, arritmogénesis y remodelación miocárdica. La investigación en genética y biología molecular seguramente encontrará recursos para la prevención y el tratamiento de las distintas cardiopatías en las que la comunicación intercelular tiene un papel fisiopatológico.


Subject(s)
Connexins/genetics , Gap Junctions , Endothelium , Molecular Biology , Myocardium
16.
Rev. argent. cardiol ; 74(2): 149-156, mar.-abr. 2006. graf
Article in Spanish | BINACIS | ID: bin-119715

ABSTRACT

Las comunicaciones intercelulares son eventos biológicos esenciales en los organismos multicelulares, asociadas con el control del crecimiento y la diferenciación celular, la apoptosis, las respuestas adaptativas de células diferenciadas y la sincronización de funciones celulares. Las uniones intercelulares, conocidas como uniones gap, estructuralmente constituidas por conexinas, tienen una participación activa en estos procesos. A nivel cardiovascular, la comunicación célula a célula es indispensable, en condiciones normales, para la embriogénesis cardíaca, la transmisión del impulso eléctrico, la sincronización de la actividad contráctil cardíaca, la transmisión de señales reflejas vasculares, entre otras funciones biológicas, mientras que en condiciones patológicas, a causa de mutaciones genéticas heredadas o adquiridas, participan en el desarrollo de cardiopatías congénitas, arritmogénesis y remodelación miocárdica. La investigación en genética y biología molecular seguramente encontrará recursos para la prevención y el tratamiento de las distintas cardiopatías en las que la comunicación intercelular tiene un papel fisiopatológico. (AU)


Subject(s)
Gap Junctions , Connexins/genetics , Molecular Biology , Myocardium , Endothelium
17.
J Immunol ; 175(9): 5885-94, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16237081

ABSTRACT

Preclinical studies demonstrated that certain cytokines are potentially useful for the induction of antitumor immune responses. However, their administration in clinical settings was only marginally useful and evoked serious toxicity. In this study, we demonstrate that the combination of autologous inactivated tumor cells expressing IL-12 and IL-10 induced tumor remission in 50-70% of mice harboring large established colon or mammary tumors and spontaneous lung metastases, with the consequent establishment of an antitumor immune memory. Mice treatment with tumor cells expressing IL-12 was only marginally effective, while expression of IL-10 was not effective at all. Administration of the combined immunotherapy stimulated the recruitment of a strong inflammatory infiltrate that correlated with local, increased expression levels of the chemokines MIP-2, MCP-1, IFN-gamma-inducible protein-10, and TCA-3 and the overexpression of IFN-gamma, but not IL-4. The combined immunotherapy was also therapeutically effective on established lung metastases from both colon and mammary tumors. The antitumor effect of the combined immunotherapy was mainly dependent on CD8+ cells although CD4+ T cells also played a role. The production of IFN-gamma and IL-4 by spleen cells and the development of tumor-specific IgG1 and IgG2a Abs indicate that each cytokine stimulated its own Th pathway and that both arms were actively engaged in the antitumor effect. This study provides the first evidence of a synergistic antitumor effect of IL-12 and IL-10 suggesting that a Th1 and a Th2 cytokine can be effectively combined as a novel rational approach for cancer immunotherapy.


Subject(s)
Colonic Neoplasms/therapy , Immunotherapy , Interleukin-10/genetics , Interleukin-12/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/therapy , Animals , Chemokines/biosynthesis , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Male , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Th1 Cells/immunology , Th2 Cells/immunology , Transfection
18.
Cancer Res ; 65(12): 5123-32, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958556

ABSTRACT

The expression of secreted protein acidic and rich in cysteine (SPARC) has been associated with the malignant progression of different types of human cancer. SPARC was associated with tumor cell capacity to migrate and invade, although its precise role in tumor progression is still elusive. In the present study, we show that SPARC produced by melanoma cells modulates the antitumor activity of polymorphonuclear leukocytes (PMN). Administration to nude mice of human melanoma cells in which SPARC expression was transiently or stably knocked down by antisense RNA (SPARC-sup cells) promoted PMN recruitment and obliterated tumor growth even when SPARC-sup cells accounted for only 10% of injected malignant cells. In addition, SPARC-sup cells stimulated the in vitro migration and triggered the antimelanoma cytotoxic capacity of human PMN, an effect that was reverted in the presence of SPARC purified from melanoma cells or by reexpressing SPARC in SPARC-sup cells. Leukotrienes, interleukin 8, and growth-related oncogene, in combination with Fas ligand and interleukin 1, mediated SPARC effects. These data indicate that SPARC plays an essential role in tumor evasion from immune surveillance through the inhibition of the antitumor PMN activity.


Subject(s)
Carrier Proteins/immunology , Melanoma/immunology , Neutrophils/immunology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , Fas Ligand Protein , Humans , Interleukin-1/immunology , Melanoma/metabolism , Membrane Glycoproteins/immunology , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Antisense/genetics , Transplantation, Heterologous
19.
J Immunol ; 171(11): 5940-7, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634105

ABSTRACT

Dendritic cells (DCs) are potent APCs and attractive vectors for cancer immunotherapy. Using the B16 melanoma, a poorly immunogenic experimental tumor that expresses low levels of MHC class I products, we investigated whether DCs loaded ex vivo with apoptotic tumor cells could elicit combined CD4(+) and CD8(+) T cell dependent, long term immunity following injection into mice. The bone marrow-derived DCs underwent maturation during overnight coculture with apoptotic melanoma cells. Following injection, DCs migrated to the draining lymph nodes comparably to control DCs at a level corresponding to approximately 0.5% of the injected inoculum. Mice vaccinated with tumor-loaded DCs were protected against an intracutaneous challenge with B16, with 80% of the mice remaining tumor-free 12 wk after challenge. CD4(+) and CD8(+) T cells were efficiently primed in vaccinated animals, as evidenced by IFN-gamma secretion after in vitro stimulation with DCs loaded with apoptotic B16 or DCs pulsed with the naturally expressed melanoma Ag, tyrosinase-related protein 2. In addition, B16 melanoma cells were recognized by immune CD8(+) T cells in vitro, and cytolytic activity against tyrosinase-related protein 2(180-188)-pulsed target cells was observed in vivo. When either CD4(+) or CD8(+) T cells were depleted at the time of challenge, the protection was completely abrogated. Mice receiving a tumor challenge 10 wk after vaccination were also protected, consistent with the induction of tumor-specific memory. Therefore, DCs loaded with cells undergoing apoptotic death can prime melanoma-specific helper and CTLs and provide long term protection against a poorly immunogenic tumor in mice.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Phagocytosis/immunology , Adoptive Transfer , Animals , Antigens, CD/biosynthesis , B7-2 Antigen , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Movement/immunology , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/biosynthesis , Immunity, Cellular , Immunity, Innate/immunology , Immunologic Memory/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Melanoma, Experimental/pathology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Up-Regulation/immunology
20.
Medicina (B.Aires) ; 54(2): 153-8, 1994. ilus, tab
Article in Spanish | LILACS | ID: lil-139581

ABSTRACT

La asociación de síndrome nefrótico con neoplasias es un hecho relativamente frecuente. Por el contrario, la insuficiencia renal rápidamente evolutiva con glomerulonefritis con semilunas extensas como consecuencia de una neoplasia es infrecuente. Presentamos el caso de un hombre de 55 años, previamente sano, que sufrió un síndrome nefrótico y una insuficiencia renal de rápida evolución en quien se detectó un cáncer pulmonar de pequeñas células. En la autopsia, se observaron proliferación mesangial y engrosamiento de las membranas basales con semilunas en el 90 por ciento de los glomérulos. La microscopia electrónica también demonstró engrosamiento de la membrana basal. No se evidenciaron estructuras compatibles con depósitos de inmunocomplejos. Realizamos una revisión de los casos comunicados de asociación de lesiones glomerulares y neoplasias y detallamos los 20 casos publicados con este tipo particular de lesión glomerular. Ninguno pertenece a esta variedad de cáncer de pulmón; los únicos tres casos de carcinoma pulmonar de pequeñas células con síndrome nefrótico referidos en la literatura se asociaron a glomerulonefritis membranosa


Subject(s)
Humans , Male , Middle Aged , Carcinoma, Small Cell/complications , Lung Neoplasms/complications , Renal Insufficiency/complications , Nephrotic Syndrome/complications , Basement Membrane/pathology , Biopsy , Carcinoma, Small Cell/pathology , Glomerulonephritis, Membranous/complications , Glomerulonephritis, Membranous/pathology , Lung Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...