Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Biol Sci ; 17(11): 2944-2956, 2021.
Article in English | MEDLINE | ID: mdl-34345218

ABSTRACT

The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.


Subject(s)
Antigens, CD/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Neoplasms, Experimental/prevention & control , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/prevention & control , Receptors, Cell Surface/immunology , Adjuvants, Immunologic , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Female , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Poly I-C/administration & dosage
2.
Cancer Chemother Pharmacol ; 86(5): 681-686, 2020 11.
Article in English | MEDLINE | ID: mdl-32980903

ABSTRACT

PURPOSE: Beta-adrenergic signaling can influence cancer progression and the use of beta blockers as adjuvant drugs in oncologic patients has been suggested. However, the involvement of beta-adrenergic blockers in tumorigenesis is poorly understood. This study investigated the action of beta-adrenergic blocker propranolol on tumor onset using a preclinical model of chemically induced oral cancer. METHODS: Thirty-two male Wistar rats were subjected to daily subcutaneous injection of beta-blocker propranolol (10 mg/kg; SubQ), while another 32 rats received only a PBS injection (sham group). One week after starting propranolol treatment, all rats were submitted to chemical induction of oral carcinogenesis with 4-nitroquinoline-1-oxide (4NQO). After 16 weeks, they were assessed for occurrence of oral squamous cell carcinoma (OSCC), in addition to measurement of tumor volume and thickness, and tissue levels of cytokines IL-6, TNF-alpha and IL-10 in the tumor microenvironment. RESULTS: Propranolol treatment reduced the occurrence of OSCC by 31%, 95% CI ( - 127, 216). Beta-adrenergic blocker significantly decreased thickness of OSCC when compared with PBS. Rats treated with propranolol exhibited a lower tumor volume when compared with control rats, but this result did not reach statistical significance. Tumors from propranolol-treated rats exhibited reduced concentrations of pro-inflammatory cytokines IL-6 and TNF-α. There was no difference in the IL-10 levels between tumors from propranolol- and sham-treated rats. CONCLUSION: Beta-adrenergic signaling may be one of the mechanisms associated with chemically induced oral carcinogenesis.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Carcinogenesis/drug effects , Mouth Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Propranolol/administration & dosage , Squamous Cell Carcinoma of Head and Neck/drug therapy , 4-Nitroquinoline-1-oxide/administration & dosage , 4-Nitroquinoline-1-oxide/toxicity , Animals , Carcinogenesis/chemically induced , Carcinogens/administration & dosage , Carcinogens/toxicity , Cytokines/immunology , Cytokines/metabolism , Disease Progression , Humans , Male , Mouth Mucosa/drug effects , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Mouth Neoplasms/chemically induced , Mouth Neoplasms/pathology , Mouth Neoplasms/prevention & control , Neoplasm Invasiveness/prevention & control , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Rats , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/chemically induced , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/prevention & control , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Front Immunol ; 9: 1914, 2018.
Article in English | MEDLINE | ID: mdl-30186285

ABSTRACT

Immunotherapy has become an important ally in the fight against distinct types of cancer. However, the metabolic plasticity of the tumor environment frequently influences the efficacy of therapeutic procedures, including those based on immunological tools. In this scenario, immunometabolic adjuvants arise as an alternative toward the development of more efficient cancer therapies. Here we demonstrated that the combination of melatonin, a neuroimmunomodulator molecule, and an indoleamine 2,3-dioxygenase (IDO) inhibitor (1-methyl-DL-tryptophan, DL-1MT) improves the efficacy of an immunotherapy (gDE7) targeting human papillomavirus (HPV)-associated tumors. Melatonin or IDO inhibitors (D-1MT and DL-1MT) directly reduced proliferation, migration, adhesion and viability of a tumor cell line (TC-1), capable to express the HPV-16 E6 and E7 oncoproteins, but could not confer in vivo antitumor protection effects. Nonetheless, combination of gDE7 with melatonin or D-1MT or DL-1MT enhanced the antitumor protective immunity of gDE7-based vaccine in mice. Notably, expression of IDO1 in stromal cells and/or immune cells, but not in tumor cells, inhibited the antitumor effects of the gDE7, as demonstrated in IDO1-deficient mice. Finally, co-administration of gDE7, melatonin and DL-1MT further improved the protective antitumor effects and the numbers of circulating E7-specific CD8+ T cells in mice previously transplanted with TC-1 cells. The unprecedented combination of melatonin and IDO inhibitors, as immunometabolic adjuvants, thus, represents a new and promising alternative for improving the efficacy of immunotherapeutic treatments of HPV-associated tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Enzyme Inhibitors/pharmacology , Human papillomavirus 16/immunology , Immunity, Cellular/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Melatonin/pharmacology , Neoplasms, Experimental/prevention & control , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/pharmacology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Human papillomavirus 16/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism
4.
Cancer Res ; 72(3): 592-603, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22139376

ABSTRACT

Toll-like receptor (TLR) ligands may be a valuable tool to promote antitumor responses by reinforcing antitumor immunity. In addition to their expression in immune cells, functional TLRs are also expressed by many cancer cells, but their significance has been controversial. In this study, we examined the action of TLR ligands on tumor pathophysiology as a result of direct tumor cell effects. B16 murine melanoma cells were stimulated in vitro with a TLR4 ligand (LPS-B16) prior to inoculation into TLR4-deficient mice (Tlr4 (lps-del)). Under such conditions, B16 cells yielded smaller tumors than nonstimulated B16 cells. The apoptosis/proliferation balance of the cells was not modified by TLR ligand treatment, nor was this effect compromised in immunocompromised nude mice. Mechanistic investigations revealed that IFNß was the critical factor produced by TLR4-activated tumor cells in mediating their in vivo outgrowth. Transcriptional analysis showed that TLR4 activation on B16 cells induced changes in the expression of type I IFN and type I IFN-related genes. Most importantly, culture supernatants from LPS-B16 cells improved the maturation of bone marrow-derived dendritic cells (BMDC) from TLR4-deficient mice, upregulating the expression of interleukin-12 and costimulatory molecules on those cells. BMDC maturation was blunted by addition of an IFNß-neutralizing antibody. Moreover, tumor growth inhibition observed in LPS-B16 tumors was abrogated in IFNAR1-deficient mice lacking a functional type I IFN receptor for binding IFN. Together, our findings show that tumor cells can be induced through the TLR4 pathway to produce IFN and positively contribute to the antitumoral immune response.


Subject(s)
Interferon-beta/immunology , Neoplasms, Experimental/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cell Line, Tumor , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
Proc Natl Acad Sci U S A ; 108(49): 19695-700, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22114198

ABSTRACT

One of the main challenges in cancer research is the development of vaccines that induce effective and long-lived protective immunity against tumors. Significant progress has been made in identifying members of the cancer testis antigen family as potential vaccine candidates. However, an ideal form for antigen delivery that induces robust and sustainable antigen-specific T-cell responses, and in particular of CD8(+) T lymphocytes, remains to be developed. Here we report the use of a recombinant nonpathogenic clone of Trypanosoma cruzi as a vaccine vector to induce vigorous and long-term T cell-mediated immunity. The rationale for using the highly attenuated T. cruzi clone was (i) the ability of the parasite to persist in host tissues and therefore to induce a long-term antigen-specific immune response; (ii) the existence of intrinsic parasite agonists for Toll-like receptors and consequent induction of highly polarized T helper cell type 1 responses; and (iii) the parasite replication in the host cell cytoplasm, leading to direct antigen presentation through the endogenous pathway and consequent induction of antigen-specific CD8(+) T cells. Importantly, we found that parasites expressing a cancer testis antigen (NY-ESO-1) were able to elicit human antigen-specific T-cell responses in vitro and solid protection against melanoma in a mouse model. Furthermore, in a therapeutic protocol, the parasites expressing NY-ESO-1 delayed the rate of tumor development in mice. We conclude that the T. cruzi vector is highly efficient in inducing T cell-mediated immunity and protection against cancer cells. More broadly, this strategy could be used to elicit a long-term T cell-mediated immunity and used for prophylaxis or therapy of chronic infectious diseases.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Membrane Proteins/immunology , Trypanosoma cruzi/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Cells, Cultured , Chagas Disease/immunology , Chagas Disease/parasitology , Chagas Disease/prevention & control , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Immunization/methods , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Transfection/methods , Trypanosoma cruzi/genetics , Trypanosoma cruzi/metabolism
6.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;44(5): 421-427, May 2011. ilus
Article in English | LILACS | ID: lil-586516

ABSTRACT

Anti-cancer DNA vaccines have attracted growing interest as a simple and non-invasive method for both the treatment and prevention of tumors induced by human papillomaviruses. Nonetheless, the low immunogenicity of parenterally administered vaccines, particularly regarding the activation of cytotoxic CD8+ T cell responses, suggests that further improvements in both vaccine composition and administration routes are still required. In the present study, we report the immune responses and anti-tumor effects of a DNA vaccine (pgD-E7E6E5) expressing three proteins (E7, E6, and E5) of the human papillomavirus type 16 genetically fused to the glycoprotein D of the human herpes simplex virus type 1, which was administered to mice by the intradermal (id) route using a gene gun. A single id dose of pgD-E7E6E5 (2 µg/dose) induced a strong activation of E7-specific interferon-γ (INF-γ)-producing CD8+ T cells and full prophylactic anti-tumor effects in the vaccinated mice. Three vaccine doses inhibited tumor growth in 70 percent of the mice with established tumors. In addition, a single vaccine dose consisting of the co-administration of pgD-E7E6E5 and the vector encoding interleukin-12 or granulocyte-macrophage colony-stimulating factor further enhanced the therapeutic anti-tumor effects and conferred protection to 60 and 50 percent of the vaccinated mice, respectively. In conclusion, id administration of pgD-E7E6E5 significantly enhanced the immunogenicity and anti-tumor effects of the DNA vaccine, representing a promising administration route for future clinical trials.


Subject(s)
Animals , Female , Mice , Cancer Vaccines/administration & dosage , /immunology , Oncogene Proteins, Viral/immunology , Simplexvirus/immunology , Vaccines, DNA/administration & dosage , Viral Envelope Proteins/immunology , /immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , /genetics , Injections, Intradermal , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/genetics , Simplexvirus/genetics , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Envelope Proteins/genetics
7.
Braz J Med Biol Res ; 44(5): 421-7, 2011 05.
Article in English | MEDLINE | ID: mdl-21445524

ABSTRACT

Anti-cancer DNA vaccines have attracted growing interest as a simple and non-invasive method for both the treatment and prevention of tumors induced by human papillomaviruses. Nonetheless, the low immunogenicity of parenterally administered vaccines, particularly regarding the activation of cytotoxic CD8+ T cell responses, suggests that further improvements in both vaccine composition and administration routes are still required. In the present study, we report the immune responses and anti-tumor effects of a DNA vaccine (pgD-E7E6E5) expressing three proteins (E7, E6, and E5) of the human papillomavirus type 16 genetically fused to the glycoprotein D of the human herpes simplex virus type 1, which was administered to mice by the intradermal (id) route using a gene gun. A single id dose of pgD-E7E6E5 (2 µg/dose) induced a strong activation of E7-specific interferon-γ (INF-γ)-producing CD8+ T cells and full prophylactic anti-tumor effects in the vaccinated mice. Three vaccine doses inhibited tumor growth in 70% of the mice with established tumors. In addition, a single vaccine dose consisting of the co-administration of pgD-E7E6E5 and the vector encoding interleukin-12 or granulocyte-macrophage colony-stimulating factor further enhanced the therapeutic anti-tumor effects and conferred protection to 60 and 50% of the vaccinated mice, respectively. In conclusion, id administration of pgD-E7E6E5 significantly enhanced the immunogenicity and anti-tumor effects of the DNA vaccine, representing a promising administration route for future clinical trials.


Subject(s)
Cancer Vaccines/administration & dosage , Human papillomavirus 16/immunology , Oncogene Proteins, Viral/immunology , Simplexvirus/immunology , Vaccines, DNA/administration & dosage , Viral Envelope Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Human papillomavirus 16/genetics , Injections, Intradermal , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/genetics , Simplexvirus/genetics , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Envelope Proteins/genetics
8.
Clin Vaccine Immunol ; 17(10): 1576-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20739505

ABSTRACT

Recombinant adenovirus or DNA vaccines encoding herpes simplex virus type 1 (HSV-1) glycoprotein D (gD) genetically fused to human papillomavirus type 16 (HPV-16) oncoproteins (E5, E6, and E7) induce antigen-specific CD8(+) T-cell responses and confer preventive resistance to transplantable murine tumor cells (TC-1 cells). In the present report, we characterized some previously uncovered aspects concerning the induction of CD8(+) T-cell responses and the therapeutic anticancer effects achieved in C57BL/6 mice immunized with pgD-E7E6E5 previously challenged with TC-1 cells. Concerning the characterization of the immune responses elicited in mice vaccinated with pgD-E7E6E5, we determined the effect of the CD4(+) T-cell requirement, longevity, and dose-dependent activation on the E7-specific CD8(+) T-cell responses. In addition, we determined the priming/boosting properties of pgD-E7E6E5 when used in combination with a recombinant serotype 68 adenovirus (AdC68) vector encoding the same chimeric antigen. Mice challenged with TC-1 cells and then immunized with three doses of pgD-E7E6E5 elicited CD8(+) T-cell responses, measured by intracellular gamma interferon (IFN-γ) and CD107a accumulation, to the three HPV-16 oncoproteins and displayed in vivo antigen-specific cytolytic activity, as demonstrated with carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled target cells pulsed with oligopeptides corresponding to the H-2D(b)-restricted immunodominant epitopes of the E7, E6, or E5 oncoprotein. Up to 70% of the mice challenged with 5 × 10(5) TC-1 cells and immunized with pgD-E7E6E5 controlled tumor development even after 3 days of tumor cell challenge. In addition, coadministration of pgD-E7E6E5 with DNA vectors encoding pGM-CSF or interleukin-12 (IL-12) enhanced the therapeutic antitumor effects for all mice challenged with TC-1 cells. In conclusion, the present results expand our previous knowledge on the immune modulation properties of the pgD-E7E6E5 vector and demonstrate, for the first time, the strong antitumor effects of the DNA vaccine, raising promising perspectives regarding the development of immunotherapeutic reagents for the control of HPV-16-associated tumors.


Subject(s)
Cancer Vaccines/immunology , Human papillomavirus 16/immunology , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/immunology , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Dose-Response Relationship, Immunologic , Female , Human papillomavirus 16/genetics , Immunization, Secondary/methods , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Time Factors , Vaccination/methods , Vaccines, DNA/administration & dosage , Viral Envelope Proteins/genetics
9.
Int J Cancer ; 124(6): 1478-82, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19065662

ABSTRACT

We tested the role of aryl hydrocarbon receptor (Ahr) gene polymorphism in the inflammatory response and in skin and lung tumorigenesis in 2 lines of mice phenotypically selected for maximum or minimum acute inflammatory reaction (AIRmax and AIRmin, respectively). Following 7,12-dimethylbenz[a]anthracene (DMBA) treatment, AIRmin but not AIRmax mice showed early skin reactions and eventually developed malignant skin tumors and lung adenocarcinomas. In skin tissue, transcript levels of IL1beta, Tnf, Il6, Tgfbeta1 and Cyp1b1 genes were upregulated in AIRmin but not AIRmax mice, consistent with the inflammatory responses to the carcinogen. These findings appeared to be related to the homozygosity status of the Ahr functional A375V polymorphism, which influences the binding capability of the receptor for DMBA: the 375A allele, encoding the high-affinity ligand-binding receptor (Ahr(b1)), segregated in AIRmin mice, whereas AIRmax mice carried the 375V, corresponding to the low-affinity binding receptor (Ahr(d)), to DMBA. The differential segregation of Ahr functional Ahr(d)versus Ahr(b1) alleles in AIRmax and AIRmin suggests a role for the Ahr gene in the control of inflammatory responsiveness and tumor development of these mouse lines.


Subject(s)
9,10-Dimethyl-1,2-benzanthracene/toxicity , Inflammation/genetics , Neoplasms, Experimental/prevention & control , Polymorphism, Genetic , Receptors, Aryl Hydrocarbon/genetics , Skin Neoplasms/chemically induced , Animals , Carcinogens/toxicity , Cytochrome P-450 CYP1A1/genetics , DNA Primers , Inflammation/chemically induced , Interferon-gamma/genetics , Interleukin-18/genetics , Interleukins/genetics , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Phenotype , Skin Neoplasms/genetics , Up-Regulation
10.
Eur J Pharmacol ; 549(1-3): 179-84, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-16996495

ABSTRACT

Recently, we demonstrated that verapamil, an L-type Ca2+ channel blocker, inhibits the activation of splenic lymphocytes during Walker 256 ascitic tumor development in adult rats. In the present study we have analyzed the changes in spleen size, splenic lymphocyte proliferation, white pulp organization and relative size as well as food intake, and levels of blood haemoglobin in Walker 256 tumor bearing rats. These rats displayed a spleen enlargement associated with a significant increase in white pulp area and TCD8+ lymphocyte proliferation. Levels of interferon-gamma, but not of interleukin-10, were elevated in tumor bearing rats, indicating a Th1-type immune response. These manifestations were accompanied by reduced food intake and anaemia. Treatment of tumor bearing rats with verapamil avoided spleen enlargement and increased expression of cytokines, as well as the splenic TCD8+ lymphocyte proliferation. In addition, verapamil treatment promoted an exacerbation of the anorexia and anaemia caused by Walker tumor development. No such effect was observed in control rats treated with verapamil. Taken together, these findings suggest that verapamil inhibits the immune response to cancer, resulting in an increase of the systemic effects induced by Walker 256 tumor.


Subject(s)
Calcium Channels/physiology , Cell Proliferation/drug effects , Neoplasms, Experimental/prevention & control , T-Lymphocytes/drug effects , Verapamil/pharmacology , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Eating/drug effects , Hemoglobins/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Male , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Organ Size/drug effects , Rats , Rats, Wistar , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Th1 Cells/drug effects , Th1 Cells/metabolism , Th1 Cells/pathology , Time Factors
11.
Microbes Infect ; 7(15): 1541-50, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16213178

ABSTRACT

DNA vaccines encoding the human papillomavirus type-16 (HPV-16) E6 and E7 oncoproteins genetically fused to the human herpes simplex virus type 1 (HSV-1) gD protein were tested in mice for induction of T cell-mediated immunity and protection against tumor cell challenge. Hybrid genes, generated after insertion of E6 or E7-encoding sequences into internal sites of the gD-encoding gene, were transcribed in vitro and the chimeric proteins were expressed at the surface of in vitro-transfected mammalian cells. Female C57BL/6 mice immunized with 4 intramuscular doses (100 microg of DNA/dose) of the DNA vaccines encoding E7 efficiently generated E7-specific CD8(+) T cells. Vaccination of mice with the DNA vaccines encoding the E7, or both E6 and E7, conferred complete protection to challenges from TC-1 tumor cells and partial therapeutic effect (40%) in mice inoculated with TC-1 cells on the same day or 5 days prior to the first vaccine dose.


Subject(s)
Herpesvirus Vaccines/immunology , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines , Repressor Proteins/immunology , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/analysis , Cytoplasm/chemistry , Disease Models, Animal , Female , Flow Cytometry , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/genetics , Human papillomavirus 16/genetics , Humans , Interferon-gamma/analysis , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Papillomavirus Infections/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Repressor Proteins/genetics , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/genetics
12.
Rev Invest Clin ; 57(3): 457-72, 2005.
Article in Spanish | MEDLINE | ID: mdl-16187707

ABSTRACT

Essential polyunsaturated fatty acids (PUFAs), linoleic acid n6 (LA) and linolenic acid (ALA) n3 obtained from the diet are precursors of the long-chain polyunsaturated fatty acids (Lc-PUFAs) arachidonic acid (AA) and docosahexaenoic acid (DHA) respectively. Consumption of PUFAs is related with a better neurological and cognitive development in newborns. It has been demonstrated that consumption of n-6 and n-3 PUFAs decreases blood triglycerides by increasing fatty acid oxidation through activation of PPARalpha or by reducing the activation of SREBP-1 inhibiting lipogenesis. Dietary PUFAs activate PPARalpha and PPARgamma increasing lipid oxidation, and decreasing insulin resistance leading in a reduction of hepatic steatosis. Beneficial effects of PUFAs have been observed in humans and in animals models of diabetes, obesity, cancer, and cardiovascular diseases. It is important to promote the consumption of PUFAs. Main food sources of PUFAs n-6 are corn, soy and safflower oil, and for PUFAs n-3 are fish, soy, canola oil and, flaxseed. Finally FAO/WHO recommends an optimal daily intake of n6/n3 of 5-10:1.


Subject(s)
Fatty Acids, Unsaturated/physiology , Adult , Aged , Aged, 80 and over , Animals , Arachidonic Acids/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , DNA-Binding Proteins/physiology , Dermatitis, Atopic/drug therapy , Diabetes Mellitus/diagnosis , Diabetes Mellitus/prevention & control , Dietary Fats/pharmacology , Dietary Fats/therapeutic use , Embryonic Development/drug effects , Embryonic Development/physiology , Fatty Acids/metabolism , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/physiology , Fatty Acids, Omega-3/therapeutic use , Fatty Acids, Omega-6/pharmacology , Fatty Acids, Omega-6/physiology , Fatty Acids, Omega-6/therapeutic use , Fatty Acids, Unsaturated/pharmacokinetics , Fatty Acids, Unsaturated/pharmacology , Fatty Acids, Unsaturated/therapeutic use , Fatty Liver/drug therapy , Female , Growth/drug effects , Growth/physiology , Humans , Hypertriglyceridemia/drug therapy , Infant, Newborn , Insulin Resistance , Lipid Peroxidation/drug effects , Male , Mice , Middle Aged , Neoplasms/drug therapy , Neoplasms/prevention & control , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/prevention & control , Obesity/drug therapy , Obesity/prevention & control , PPAR alpha/agonists , PPAR gamma/agonists , Plant Oils/pharmacology , Plant Oils/therapeutic use , Pregnancy , Rats
13.
Int J Mol Med ; 16(4): 565-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142388

ABSTRACT

Bacillus Calmette-Guérin (BCG) is considered to be one of the most effective treatments for superficial and in situ bladder cancer. However, either failure to respond initially or relapse within the first 5 years of treatment has been observed in some patients. As nitric oxide (NO) has been detected in the bladder of BCG-treated patients, we analyzed the role of endogenous NO generated after BCG treatments on human (T24) and murine (MB49 and MBT2) bladder tumor cells in the viability of tumor and immune cells, both in vitro and in vivo. In vitro inhibition of cancer cells after BCG treatment was evaluated by cell titer assay. NO production was determined as nitrite by Griess reagent. The death of immunocytes was evaluated by 51Cr release. Tumor histology with hematoxylin and eosin and Masson's trichrome staining was performed. BCG induced a direct inhibition of tumor cell growth in vitro, independently of NO levels. Besides, BCG-mediated NO production by tumor cells induced the death of spleen and peritoneal cells in syngeneic mice. The in vivo inhibition of NO synthase (NOS) activity by NG-nitro-L-arginine methyl ester in combination with BCG, improved tumor regression by generating a healing tissue. The increase of NO generated after BCG administration may induce the death of immunocytes. The in vivo inhibition of NO ameliorated immunotherapy with BCG by additional tumor growth inhibition. Our results suggested the possibility that the final outcome of patients with bladder tumors may improve by modulating NOS activity concomitantly with BCG therapy.


Subject(s)
BCG Vaccine/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Urinary Bladder Neoplasms/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Injections, Intralesional , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/therapeutic use , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/pharmacology , Spleen/cytology , Spleen/drug effects , Time Factors , Treatment Outcome , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
14.
Braz J Med Biol Res ; 37(7): 1029-34, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15264010

ABSTRACT

Chemoprotection by dietary agents is a promising strategy for cancer prevention. The aim of the present study was to evaluate the combined effect of tomato and garlic against 7,12-dimethylbenz-[a]anthracene (DMBA)-induced genetic damage and oxidative stress in 12-14-week-old male Swiss albino mice. The animals were randomized into experimental and control groups and divided into eight groups of five animals each. Group 1 animals were injected intraperitoneally with 35 mg/kg body weight DMBA suspended in peanut oil as a single dose. Groups 2-4 animals received tomato (500 mg/kg body weight), garlic (125 mg/kg body weight) and a combination of tomato and garlic for 5 days by gavage, respectively, followed by DMBA 1.5 h after the final feeding. The doses of tomato and garlic correspond to the average human daily consumption. Animals in groups 5, 6 and 7 received tomato alone, garlic alone and tomato + garlic combination, respectively, for 5 days. Group 8 animals received the same volume of water and served as control. The incidence of bone marrow micronuclei and the extent of lipid peroxidation and the concentrations of antioxidants glutathione, glutathione peroxidase and glutathione-S-transferase were measured in the liver, 48 h after DMBA exposure. Increased frequency of micronuclei and enhanced lipid peroxidation accompanied by compromised antioxidant defenses were observed in DMBA-treated animals. Although pretreatment with tomato or garlic significantly reduced the frequency of DMBA-induced bone marrow micronuclei, the combination of tomato and garlic exhibited more profound effect in inhibiting DMBA-induced genotoxicity and oxidative stress. We suggest that a broad spectrum of antimutagenic and anticlastogenic effects can be achieved through an effective combination of functional foods such as tomato and garlic.


Subject(s)
DNA Damage/drug effects , Garlic , Lipid Peroxidation/drug effects , Neoplasms, Experimental/prevention & control , Oxidative Stress , Solanum lycopersicum , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogens , Glutathione/analysis , Glutathione Peroxidase/analysis , Glutathione Transferase/analysis , Male , Mice , Micronucleus Tests , Plant Extracts/administration & dosage , Random Allocation
15.
Cancer Lett ; 190(1): 45-50, 2003 Feb 10.
Article in English | MEDLINE | ID: mdl-12536076

ABSTRACT

The influence of myenteric denervation on the development of gastric tumors induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was studied after chemical denervation of the rat stomach with benzalkonium chloride (BAC). Three groups were evaluated: control, denervated and denervated with pyloroplasty. Random bred male Wistar rats were given MNNG in drinking water (100 mg/l) for 28 weeks. After the sacrifice of animals, the stomachs were removed for morphological study. BAC reduced myenteric neurons number, increased the gastric mucosa area and decreased the adenocarcinomas number and size. This decrease was more evident when denervation was associated with pyloroplasty. These results indicate that myenteric denervation reduces the incidence of experimentally induced gastric tumors.


Subject(s)
Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control , Sympathectomy, Chemical , Animals , Benzalkonium Compounds/pharmacology , Carcinogens , Male , Methylnitronitrosoguanidine , Neoplasms, Experimental/prevention & control , Rats , Rats, Wistar , Stomach/innervation
16.
Vopr Onkol ; 48(6): 710-3, 2002.
Article in Russian | MEDLINE | ID: mdl-12530270

ABSTRACT

Inhibition of experimental tumor growth, life-span, mitotic activity and cytologic damage have been studied in cells of ascitic and solid carcinoma of Ehrlich in the course of biotherapy with vaccines of Venezuelan encephalomyelitis virus, mumps and influenza viruses. Summing up, it is concluded that the vaccine of Venezuelan encephalomyelitis virus looks most promising as far as further research in development of an antitumor viral drug is concerned.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Encephalitis Virus, Venezuelan Equine/immunology , Influenza Vaccines/therapeutic use , Mumps Vaccine/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Viral Vaccines/therapeutic use , Animals , Female , Male , Mice , Neoplasms, Experimental/prevention & control , Neoplasms, Experimental/virology
17.
Eur J Cancer Prev ; 7(4): 337-42, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9806123

ABSTRACT

Hepatocarcinogenesis (HC) induced by various carcinogens such as 1,4-dimethylaminoazobenzene (DAB) is a multistep and complex process. The anticancer efficacy of beta-carotene (beta C) was evaluated by estimating some biochemical parameters during the initiation stage of HC. beta C dietary supplementation partially prevented the rise in delta-aminolevulinate synthetase activity. P 450 levels were dramatically enhanced in all groups studied. beta C administration did not overcome catalase inactivation due to DAB treatment; however, superoxide dismutase activity levels showed to be less decreased in the DAB + beta C animals in comparison to the DAB group. The great enhancement provoked by DAB of glutathione S-transferase, a proposed marker of HC, was partially reversed by beta C. In conclusion, heme pathway regulation, drug metabolism, and natural oxidative defence systems, strikingly modified in DAB fed animals, were partially controlled by provitamin A. The potential use of beta C in preventing carcinogenesis is suggested.


Subject(s)
Antioxidants/pharmacology , Carcinogens/toxicity , Neoplasms, Experimental/prevention & control , beta Carotene/pharmacology , p-Dimethylaminoazobenzene/toxicity , Animals , Antioxidants/therapeutic use , Drug Antagonism , Male , Mice , beta Carotene/therapeutic use
18.
Salud Publica Mex ; 39(5): 451-62, 1997.
Article in Spanish | MEDLINE | ID: mdl-9424727

ABSTRACT

Several genetic alterations occur during the transformation process from normal to tumor cells, that involve the loss of fidelity of processes as replication, reparation, and segregation of the genomic material. Although normal cells have defense mechanisms against cancer progression, in tumor cells different escape pathways are activated leading to tumor progression. Recent advances have permitted cancer research to focus on the identification of some of its etiological factors. The knowledge of cell cycle reveals a precise mechanism achieved by the coordinated interactions and functions of cyclin-dependent kinases, control checkpoint, and repair pathways. Furthermore, it has been demonstrated that this coordinated function can be abrogated by specific genetic changes. These findings suggest that the molecular mechanisms responsible for cellular transformation may help to identify potential targets to improve cancer therapies.


Subject(s)
Cell Transformation, Neoplastic/pathology , Neoplasms, Experimental/drug therapy , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Genes, Retinoblastoma/physiology , Humans , Neoplasms/etiology , Neoplasms/prevention & control , Neoplasms, Experimental/etiology , Neoplasms, Experimental/prevention & control , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/physiology
19.
Rev Med Chil ; 119(4): 458-64, 1991 Apr.
Article in Spanish | MEDLINE | ID: mdl-1842994

ABSTRACT

In experimental animals, restriction of caloric intake is associated to a decreased incidence in the number and size of both spontaneous and induced tumors. Epidemiologic evidence of a similar effect in humans is not conclusive. However, some dietary recommendations regarding calories, protein and fiber content may be made in order to help prevent cancer development in human subjects.


Subject(s)
Energy Intake/physiology , Neoplasms/prevention & control , Animals , Female , Humans , Male , Neoplasms, Experimental/prevention & control , Rats
SELECTION OF CITATIONS
SEARCH DETAIL