Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cell Immunol ; 367: 104394, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34198057

RESUMEN

Melanomas exhibit the highest rate of heterogeneity among cancer cell types. In this study, we tested the two types of B16 melanoma cells (B16-S0-1 and B16-S1-1) showing resistance to antitumor immunity. These cells expressed Trp2 protein. Contrary to B16 and B16-S0-1 cells, B16-S1-1 cells failed to stimulate IFN-γ responses in Trp2-specific CD8+ T cells, suggesting that B16-S1-1 cells may have lost the ability to present antigen to Ag-specific CTLs in the context of MHC class I molecules. However, B16-S0-1 cells exhibited active Stat3 and decreased Bcl-2 expression, which were found to be not associated with immune escape. B16-S0-1 cells were more resistant to granzyme B-mediated caspase activation and apoptosis than B16 cells. Thus, these data show that B16 cells escape antitumor immune responses through the loss of epitope presentation to CTLs and the acquisition of tumor cell resistance to granzyme B-mediated caspase activation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/metabolismo , Melanoma/inmunología , Animales , Presentación de Antígeno , Apoptosis , Citotoxicidad Inmunológica , Epítopos de Linfocito T/inmunología , Femenino , Granzimas/metabolismo , Evasión Inmune , Interferón gamma/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos
2.
J Immunol ; 196(5): 2410-23, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26829987

RESUMEN

A long-standing question in the field of tumor immunotherapy is how Th2 cytokines block tumor growth. Their antitumor effects are particularly prominent when they are secreted continuously in tumors, suggesting that Th2 cytokines may create a tumor microenvironment unfavorable for tumor growth independently of adaptive immunity. In this study, we show that local production of IL-33 establishes a high number of type 2 innate lymphoid cells (ILC2s) with potent antitumor activity. IL-33 promotes secretion of a massive amount of CXCR2 ligands from ILC2s but creates a tumor microenvironment where tumor cells express CXCR2 through a dysfunctional angiogenesis/hypoxia/reactive oxygen species axis. These two signaling events converge to reinforce tumor cell-specific apoptosis through CXCR2. Our results identify a previously unrecognized antitumor therapeutic pathway wherein ILC2s play a central role.


Asunto(s)
Inmunidad Innata , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Antígenos de Superficie/metabolismo , Biomarcadores , Línea Celular Tumoral , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoxia/metabolismo , Inmunofenotipificación , Interleucina-33/metabolismo , Linfocitos/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Carga Tumoral , Microambiente Tumoral
3.
Cancer Immunol Immunother ; 61(10): 1671-82, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22382361

RESUMEN

Intratumoral electroporation (IT-EP) with IL-12 cDNA (IT-EP/IL12) can lead to the eradication of established B16 melanoma tumors in mice. Here, we explore the immunological mechanism of the antitumor effects generated by this therapy. The results show that IT-EP/IL12 applied only once resulted in eradication in 70% animals with large established B16 tumors. Tumor eradication required the participation of CD8+ T cells, but not CD4+ T cells and NK cells. IT-EP/IL12 induced antigen-specific CD8+ T cell responses against the immunodominant Trp2(180-188) epitope and generated a systemic response, resulting in significant therapeutic effects against distal, untreated tumors. The therapeutic effect of IT-EP/IL12 was absent in perforin-deficient mice, indicating that tumor elimination occurred through conventional perforin/granzyme lysis by CTLs. Moreover, this therapy induced some degree of immunological memory that protected approximately one-third of the cured mice against a subsequent tumor challenge. Moreover, antitumor efficacy and long-term protection against B16 were significantly improved by concurrent Trp2 peptide immunization through more induction of Ag-specific CTL responses and more attraction of IFN-γ-expressing CD8+ T cells into tumor sites. The antitumor effect of IT-EP/IL12 required the participation of IFN-γ, which was shown to induce MHC class I expression on B16 cells and increase the lytic activity of the CD8+ CTL generated by IT-EP/IL12. The results from these animal studies may help in the development of IT-EP/IL12 for cancer patients.


Asunto(s)
Electroquimioterapia/métodos , Granzimas/inmunología , Interferón gamma/inmunología , Interleucina-12/genética , Oxidorreductasas Intramoleculares/inmunología , Melanoma Experimental/terapia , Proteínas Citotóxicas Formadoras de Poros/inmunología , Neoplasias Cutáneas/terapia , Linfocitos T Citotóxicos/inmunología , Animales , ADN Complementario/genética , Modelos Animales de Enfermedad , Femenino , Memoria Inmunológica , Interleucina-12/inmunología , Ratones
4.
J Minim Invasive Gynecol ; 18(2): 157-63, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21216677

RESUMEN

STUDY OBJECTIVE: To estimate whether additional bleeding control can be safely achieved during laparoscopic myomectomy using bipolar electrosurgery over the suture sites in patients with blood oozing despite sufficient myometrial sutures. DESIGN: Retrospective case control study (Canadian Task Force classification II-1). SETTING: University teaching hospital. PATIENTS: One hundred twenty-six women who underwent laparoscopic myomectomy performed by one surgeon. INTERVENTIONS: Changes in maximum tensile strength of various suture materials were measured at tensinometry after bipolar electrosurgery or diathermy. Bipolar electrosurgery was performed over suture sites during laparoscopic myomectomy after adequate suturing (bipolar group, n = 64). Clinical outcomes were compared with those in matched controls (control group, n = 62]. MEASUREMENTS AND MAIN RESULTS: Polyglactin 910 (Vicryl) and glycolide-lactide copolymer (Polysorb) sutures exhibited no substantial changes in maximum tensile strength after 2 seconds of bipolar electrosurgery. However, both sutures demonstrated a decrease in maximum tensile strength of 43.5% and 17.4%, respectively, after 4 seconds of bipolar electrosurgery at 40 W. Compared with the control group, in the bipolar group mean (SD) postoperative hemoglobin concentration was higher (11.1 [1.3] g/dL vs 10.5 [1.3] g/dL), total drainage volume was less (244.6 [133.7] mL vs 380.2 [196.0] mL), a drain was required for a shorter time (2.0 [0.7] days vs 2.8 [0.7] days), and hospital stay was shorter (4.3 [1.6] days vs 5.3 [1.7] days) (p <.05). CONCLUSIONS: During laparoscopic myomectomy, additional bleeding control can be achieved by using careful short duration bipolar electrosurgery over the suture site. However, application of excessive bipolar electrosurgery (>40 W and ≥ 4 seconds) tends to weaken suture material.


Asunto(s)
Electrocirugia/métodos , Laparoscopía/métodos , Útero/cirugía , Estudios de Casos y Controles , Femenino , Humanos , Poliglactina 910 , Estudios Retrospectivos , Suturas , Resistencia a la Tracción
5.
Hum Vaccin Immunother ; 17(7): 1910-1922, 2021 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-33522416

RESUMEN

Anti-programmed death ligand 1 (PD-L1) therapy has been beneficial in treating patients with certain cancers. Here, we tested whether anti-PD-L1 therapy is effective for controlling different types of tumors using animal models of TC-1, MC38 and B16. We found that, despite PD-L1 expression, anti-PD-L1 therapy showed little and some antitumor activity in the TC-1 and MC38 models. However, anti-PD-L1 therapy exhibited a more dramatic antitumor effect in the B16 model. This difference in antitumor responses was likely associated with the CD8 + T cell infiltration status of tumor tissues. In the B16 model, CD8 + T cells and to a lesser degree NK cells were found to be responsible for the antitumor response of anti-PD-L1 therapy, as determined by immune cell subset depletion. In particular, CD8 + T cells from B16-bearing mice produced an IFN-γ in response to B16 cells and citrate phosphate buffer-treated B16 cell peptide elutes but not to an immunodominant class I epitope, Trp2180-188, suggesting that CD8 + T cells that recognize neoantigens were induced in B16 tumor-bearing mice and then reactivated by anti-PD-L1 for tumor control. When B16 tumor-bearing mice were treated with anti-PD-L1 in combination with Trp2180-188 peptide vaccines, they displayed significantly more tumor control than either single therapy. Taken together, these studies show that B16 melanomas are more effectively controlled through reactivation of tumor-infiltrating lymphocytes by anti-PD-L1 therapy. Moreover, combined therapy using anti-PD-L1 and Trp2 peptide vaccines is more beneficial for controlling B16 melanomas through reactivation of neoantigen-specific CD8 + T cells and induction of Trp2-specific CD8 + T cells.


Asunto(s)
Vacunas contra el Cáncer , Melanoma Experimental , Animales , Antígeno B7-H1 , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Células Asesinas Naturales , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Péptidos
6.
Mol Ther ; 17(5): 906-13, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19277009

RESUMEN

Adoptive cytotoxic T lymphocyte (CTL) therapy has an important implication in treating cancer patients. Here, we investigate whether adoptive transfer of human papillomavirus (HPV) E7-specific CTL can enhance tumor chemoresponse using an established cervical cancer animal model. Cisplatin-based chemotherapy plus CTL therapy showed an improved therapeutic effectiveness, along with antitumor protective responses to a parental tumor cell rechallenge. Cisplatin treatment dose-dependently increased the expression of Fas, intercellular adhesion molecule (ICAM)-1, and major histocompatibility complex (MHC) class I antigens (Ags) on tumor cells in vitro. However, CTL-expressing FasL failed to improve antitumor activity in vitro and in animals, resulting from nonfunctional Fas expressed on tumor cells. In contrast, ethylene glycol tetraacetic acid (EGTA) treatment blocked increased sensitivity of cisplatin-treated tumor cells to CTL-mediated killing in vitro, suggesting an important role of the perforin/granzyme-mediated pathway for improved therapeutic effectiveness. This notion was further confirmed by perforin knockout animal studies. Thus, this study shows that (i) modulation of Ag (Fas, ICAM-1) expression by tumor cells has little effect on their increased sensitivity to CTL-mediated killing, (ii) improved therapeutic effectiveness is mediated mainly through the perforin/granzyme-mediated tumor killing pathway, and (iii) a combination of chemotherapy and adoptive E7-specific CTL transfer augments antitumor therapeutic activity in vivo. This finding may have important implications for treating HPV-associated cervical cancer.


Asunto(s)
Granzimas/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Perforina/metabolismo , Linfocitos T Citotóxicos/metabolismo , Animales , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Cisplatino/farmacología , Ácido Egtácico/farmacología , Femenino , Citometría de Flujo , Granzimas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Perforina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Citotóxicos/efectos de los fármacos
7.
Front Immunol ; 11: 604413, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33363545

RESUMEN

Legionella pneumophila, an intracellular bacterium, may cause life-threatening pneumonia in immunocompromised individuals. Mononuclear cells and antibodies have been reported to be associated with the host defense response against L. pneumophila. This study is to determine whether Legionella peptidoglycan-associated lipoprotein (PAL)-specific CD8+ T cells are directly associated with protection against L. pneumophila, with a focus on potential epitopes. Synthetic peptides derived from PAL of L. pneumophila were obtained and tested through in vitro and in vivo cytotoxic T lymphocyte (CTL) assays for immunogenicity. PAL DNA vaccines or a peptide epitope with or without CpG-oligodeoxynucleotides (ODN) was evaluated for protection against L. pneumophila infection in animal models. When mice were immunized with DNA vaccines expressing the PAL of L. pneumophila, they were significantly protected against a lethal challenge with L. pneumophila through induction of antigen-specific CD8+ CTLs. Of the 13 PAL peptides tested, PAL92-100 (EYLKTHPGA) was the most immunogenic and induced the strongest CTL responses. When mice were immunized with the PAL92-100 peptide plus CpG-ODN, they were protected against the lethal challenge, while control mice died within 3-6 days after the challenge. Consistent with lung tissue histological data, bacterial counts in the lungs of immunized mice were significantly lower than those in control mice. Also, the amino acid sequence of PAL92-100 peptides is conserved among various Legionella species. To our knowledge, this study is the first to demonstrate that PAL92-100-specific CD8+ T cells play a central role in the host defense response against L. pneumophila.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Epítopos , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/prevención & control , Pulmón/inmunología , Fragmentos de Péptidos/administración & dosificación , Proteoglicanos/administración & dosificación , Linfocitos T Citotóxicos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Carga Bacteriana , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Células Cultivadas , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Inmunización , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/metabolismo , Enfermedad de los Legionarios/microbiología , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Activación de Linfocitos , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Proteoglicanos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/microbiología
8.
Immunology ; 128(1 Suppl): e707-17, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19740332

RESUMEN

Interleukin-12 (IL-12) has been shown to enhance cellular immunity in vitro and in vivo. The beneficial roles of IL-12 as a DNA vaccine adjuvant have been commonly observed. Here the impact of IL-12 complementary DNA (cDNA) as an adjuvant for a human papillomavirus (HPV) type 16 E7 DNA vaccine is investigated in a mouse tumour model. Coinjection of E7 DNA vaccine with IL-12 cDNA completely suppressed antigen-specific cytotoxic T-lymphocyte (CTL) responses, leading to a complete loss of antitumour protection from a tumour cell challenge. In addition, antigen-specific antibody and T helper cell proliferative responses were also suppressed by IL-12 cDNA coinjection. This inhibition was observed over different IL-12 cDNA doses. Furthermore, separate leg injections of IL-12 and E7 cDNAs suppressed antigen-specific CTL and tumour protective responses, but not antibody and T helper cell proliferative responses, suggesting different pathways for suppression of these two separate responses. Further knockout animal studies demonstrated that interferon-gamma and nitric oxide are not directly associated with suppression of antigen-specific antibody responses by IL-12 cDNA coinjection. However, nitric oxide was found to be involved in suppression of antigen-specific CTL and tumour protective responses by IL-12 cDNA coinjection. These data suggest that coinjection of IL-12 cDNA results in suppression of E7-specific CTL responses through nitric oxide, leading to a loss of antitumour resistance in this DNA vaccine model. This study further shows that the adjuvant effect of IL-12 is dependent on the antigen types tested.


Asunto(s)
Papillomavirus Humano 16/inmunología , Interleucina-12/inmunología , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Linfocitos T Citotóxicos/inmunología , Neoplasias del Cuello Uterino/prevención & control , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , ADN Complementario/administración & dosificación , ADN Complementario/genética , ADN Complementario/inmunología , Femenino , Papillomavirus Humano 16/genética , Humanos , Inmunidad Celular , Inmunidad Humoral , Interferón gamma/biosíntesis , Interleucina-12/genética , Ratones , Ratones Endogámicos C57BL , Ácido Nítrico/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Neoplasias del Cuello Uterino/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
9.
Sci Rep ; 9(1): 2855, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814560

RESUMEN

Tumor cells tend to behave differently in response to immune selective conditions. Contrary to those in therapeutic antitumor conditions, tumor cells in prophylactic antitumor conditions lose antigen expression for antitumor immune escape. Here, using a CT26/HER2 tumor model, we investigate the underlying mechanism(s). We selected tumor cell variants (CT26/HER2-A1 and -A2) displaying resistance to antitumor protective immunity and loss of HER2 antigen expression. These immune-resistant cells failed to induce Ag-specific IgG and IFN-γ responses while forming tumors at the same rate as CT26/HER2 cells. RT-PCR, qRT-PCR, PCR, Western blot and DNA sequencing analyses demonstrated that HER2 expression was inhibited at the post-transcriptional level in these immune-resistant cells, suggesting that tumor cells may escape antitumor immunity through the post-transcriptional regulation of antigen gene expression. The proteasome and lysosomal protein degradation pathways were not responsible for antigen loss, as determined by an inhibitor assay. Finally, HER2 mRNA was found to be not present in the monosomes and polysomes of CT26/HER2-A2 cells, as opposed to CT26/HER2 cells, suggesting that the translation activity of HER2 mRNAs may be suppressed in these immune-resistant cells. Taken together, our results report a new mechanism by which tumor cells respond to antitumor protective immunity for antitumor immune evasion.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Experimentales/inmunología , ARN Mensajero/inmunología , ARN Neoplásico/inmunología , Receptor ErbB-2/inmunología , Escape del Tumor , Animales , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , ARN Mensajero/genética , ARN Neoplásico/genética , Receptor ErbB-2/genética
10.
Oncol Rep ; 19(4): 1027-32, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18357392

RESUMEN

The proteasome plays a pivotal role in controlling cell proliferation, apoptosis, and differentiation in a variety of tumor cells. Bortezomib is a boronic acid dipeptide derivative, which is a selective and potent inhibitor of the proteasome and has prominent effects in vitro and in vivo against several solid tumors. We examined the anti-proliferative and apoptotic effects of bortezomib in three gastric cancer cell lines (SNU638, MUGC-3 and MKN-28), along with its antitumor combination effects with other chemotherapeutic agents. Tumor cell growth inhibition and apoptosis was measured by MTT assay and FACS analysis, respectively. Apoptosis- and cell cycle-associated protein expression levels were measured by Western blot assay. Bortezomib induced the suppression of tumor cell growth and apoptosis in a dose-dependent manner with an inhibitory dose (ID)50 of approximately 0.5 microg/ml in all gastric cancer cell lines tested. Further combination treatment with cisplatin and docetaxel, in particular with docetaxel displayed dramatically increased tumor cell growth suppression in all three gastric cancer cell lines, as compared to single drug treatment alone. This was concomitant with the induction patterns of apoptotic cells. Bortezomib treatment increased the Bax protein expression. Moreover, combination treatment of bortezomib plus docetaxel resulted in a dramatic increase in the Bax expression. In contrast, Bcl-2 expression was decreased by combination treatment with bortezomib plus docetaxel in SNU638 cells. Finally, bortezomib, docetaxel and to a greater degree bortezomib plus docetaxel increased the expression levels of p27 proteins even without influencing p53 expression levels. Bortezomib has profound effects on tumor cell growth inhibition and induction of apoptosis in human gastric cancer cells, suggesting that bortezomib may be an effective therapeutic drug for patients with gastric cancer. Further combination studies with other chemotherapeutic drugs, in particular docetaxel showing more tumor cell growth inhibition and apoptosis suggest that combining bortezomib with docetaxel might be more effective for displaying tumor cell growth inhibitory effects in gastric cancer cells through regulation of Bcl-2, Bax and p27 proteins in vitro.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácidos Borónicos/farmacología , Inhibidores de Proteasoma , Pirazinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Bortezomib , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/análisis , Docetaxel , Humanos , Neoplasias Gástricas/química , Neoplasias Gástricas/patología , Taxoides/farmacología , Proteína p53 Supresora de Tumor/análisis
11.
Clin Cancer Res ; 13(1): 341-9, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17200373

RESUMEN

PURPOSE: The goal of this study was to investigate the therapeutic potentials of combining chemotherapy with human papillomavirus (HPV) E7 subunit vaccines in an animal tumor model and to determine the underlying therapeutic mechanisms. EXPERIMENTAL DESIGN: Animals bearing HPV E6/E7-expressing tumors were treated intratumorally with a selected cytotoxic drug, cisplatin, twice at 1-week interval and s.c. with E7 subunit vaccines thrice at 1-week interval. Tumor chemoimmunoresponse was measured by tumor size. Ag-specific CTL activities and tumor histology were checked in mice under treatments. Apoptosis, in vivo T-cell subset depletion, adoptive CTL transfer, and tumor regression were used to determine the mechanisms for antitumor therapeutic effects. RESULTS: Combined therapy using cisplatin plus E7 subunit vaccines improved cure and recurrence rates of tumors and long-term antitumor immunity dramatically more than single therapy alone. In particular, both components of E7 subunit vaccines were required for induction of Ag-specific CTL as well as therapeutic synergy when combined with cisplatin. This therapeutic synergy was abrogated by depletion of CD8(+) T cells in vivo and was concomitant with histologic changes (such as heavy infiltration of lymphocytes and reduced tumor cell density). Finally, the increased sensitivity of cisplatin-treated tumors to CTL-mediated killing was found to be responsible for therapeutic synergy. CONCLUSIONS: E7 subunit vaccines plus cisplatin mediate antitumor therapeutic synergy through the increased sensitivity of cisplatin-treated tumors to CTL-mediated killing. Moreover, E7-based therapeutic vaccines have the potential to improve chemotherapy in patients with cervical cancer.


Asunto(s)
Vacunas contra el Cáncer , Cisplatino/administración & dosificación , Terapia Combinada , Proteínas E7 de Papillomavirus/química , Vacunas contra Papillomavirus/metabolismo , Linfocitos T Citotóxicos/citología , Animales , Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
12.
Clin Exp Vaccine Res ; 7(2): 119-128, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30112351

RESUMEN

PURPOSE: The goal of this study was to purify and characterize Ebola virus glycoprotein (GP)-specific IgG antibodies from hybridoma clones. MATERIALS AND METHODS: For hybridoma production, mice were injected by intramuscular-electroporation with GP DNA vaccines, and boosted with GP vaccines. The spleen cells were used for producing GP-specific hybridoma. Enzyme-linked immunosorbent assay, Western blot assay, flow cytometry, and virus-neutralizing assay were used to test the ability of monoclonal IgG antibodies to recognize GP and neutralize Ebola virus. RESULTS: Twelve hybridomas, the cell supernatants of which displayed GP-binding activity by enzyme-linked immunosorbent assay and the presence of both IgG heavy and light chains by Western blot assay, were chosen as a possible IgG producer. Among these, five clones (C36-1, D11-3, D12-1, D34-2, and E140-2) were identified to secrete monoclonal IgG antibodies. When the monoclonal IgG antibodies from the 5 clones were tested for their antigen specificity, they recognized GP in an antigen-specific and IgG dose-dependent manner. They remained reactive to GP at the lowest tested concentrations (1.953-7.8 ng/mL). In particular, IgG antibodies from clones D11-3, D12-1, and E140-2 recognized the native forms of GP expressed on the cell surface. These antibodies were identified as IgG1, IgG2a, or IgG2b kappa types and appeared to recognize the native forms of GP, but not the denatured forms of GP, as determined by Western blot assay. Despite their GP-binding activity, none of the IgG antibodies neutralized Ebola virus infection in vitro, suggesting that these antibodies are unable to neutralize Ebola virus infection. CONCLUSION: This study shows that the purified IgG antibodies from 5 clones (C36-1, D11-3, D12-1, D34-2, and E140-2) possess GP-binding activity but not Ebola virus-neutralizing activity.

13.
DNA Cell Biol ; 37(10): 850-860, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30227079

RESUMEN

The role of chemotherapeutic agents in tumor immunotherapy is still controversial. In this study, we test using a TC-1 tumor model whether gemcitabine plus E7 peptide vaccine regimens (E7 peptides+CpG-ODN+anti-4-1BB Abs) may result in tumor cure in mice with large established tumors, with a focus on their effects on Ag-specific cytotoxic T lymphocyte (CTL) and myeloid-derived suppressor cell levels. Gemcitabine inhibited tumor growth by its direct cytotoxicity to tumor cells in vivo. E7 peptide vaccine regimens enhanced Ag-specific CTL lytic and antitumor therapeutic activity. Initial combination therapy using gemcitabine and E7 peptide vaccine regimens resulted in tumor regression with tumor relapse in animals with large established tumors, which appeared to result from the suppression of Ag-specific CTL activity by gemcitabine treatment. However, optimization of gemcitabine therapy by reducing its dose and frequency led to complete tumor regression without any recurring tumors in all tested mice even after discontinuation of therapy, possibly due to Ag-specific CTL responses. Thus, this study shows that the optimal dose and therapy frequency of gemcitabine are critical for achieving tumor cure in tumor-bearing animals undergoing E7 peptide vaccine regimen therapy, mainly by preventing CTL suppression. These findings may have implications for designing peptide-based therapeutic vaccines in cancer patients undergoing chemotherapy.


Asunto(s)
Adenocarcinoma/terapia , Antimetabolitos Antineoplásicos/farmacología , Vacunas contra el Cáncer/administración & dosificación , Terapia Combinada/métodos , Desoxicitidina/análogos & derivados , Linfocitos T Citotóxicos/efectos de los fármacos , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Anticuerpos/farmacología , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/química , Vacunas contra el Cáncer/química , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Desoxicitidina/farmacología , Quimioterapia/métodos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Humanos , Inmunización , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/síntesis química , Péptidos/administración & dosificación , Péptidos/síntesis química , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Carga Tumoral/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Gemcitabina
14.
Biotechnol Lett ; 29(4): 511-6, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17235489

RESUMEN

An attenuated strain of Salmonella typhimurium has been tested in animals and clinically as an anticancer agent due to its in vivo tumor-targeting and tumoricidal properties. We exploited a genetically-engineered S. typhimurium harboring Flt3 Ligand (Flt3L) expression vectors as a tumoricidal agent to enhance its therapeutic efficacy. Flt3L showed tumoricidal effects when expressed in tumor cells in vitro. When melanoma-bearing mice were treated locally with Salmonella, S. typhimurim with Flt3L expression vectors inhibited tumor growth more than Salmonella controls (50% vs. 0% in tumor regression rates). Moreover, it prolonged survivals of animals without induction of memory antitumor protective responses to a parental tumor re-challenge (50% vs. 0% in survival rates). These results suggest that a genetically engineered S. typhimurium with Flt3L expression vectors has the potential to be applicable as a safer and more effective tumor-targeting and tumoricidal agent.


Asunto(s)
ADN Bacteriano/administración & dosificación , Melanoma/patología , Melanoma/terapia , Proteínas de la Membrana/genética , Plásmidos/administración & dosificación , Salmonella typhimurium/genética , Transfección/métodos , Animales , Línea Celular Tumoral , Terapia Genética/métodos , Vectores Genéticos/genética , Melanoma/genética , Proteínas de la Membrana/uso terapéutico , Ratones , Ratones Endogámicos C57BL
15.
DNA Cell Biol ; 36(9): 801-811, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28777668

RESUMEN

Therapeutic control of tumors is challenging as they tend to alter their biological functions and microenvironment. In a CT26/HER2 tumor model, HER2 DNA vaccines and even anti-PD-L1 Abs failed to display antitumor therapeutic activity while inducing Ag-specific cytotoxic T lymphocyte (CTL) activity. To clarify this contradictory finding, we selected tumor cells (CT26/HER2-1) from one tumor-bearing animal in the therapeutic model. CT26/HER2-1 cells behaved similar to wild-type CT26/HER2 cells in their HER2 expression, immune cell stimulation for IFN-γ production, and antitumor immune sensitivity. A similar finding was obtained with additional CT26/HER2-2, -3, -4, -5, and -6 cells from the therapeutic model, suggesting that a lack of antitumor therapeutic activity of HER2 DNA vaccines might be ascribed to a factor in the tumor microenvironment, but not to an alteration in tumor cell functions. When tumor-bearing mice were depleted of myeloid-derived suppressor cells (MDSCs) by anti-Gr-1 Ab treatment, they displayed HER2 vaccine-mediated antitumor activity, suggesting a role of MDSCs in blocking antitumor activity. Moreover, when tumor-bearing mice were treated with gemcitabine, they displayed HER2 vaccine-mediated antitumor activity, suggesting that cytotoxic drug treatment makes tumor cells susceptible to lysis by CTLs. Thus, these studies show that therapeutic control of HER2 DNA vaccines can be achieved by anti-Gr-1 Ab treatment through MDSC depletion and by gemcitabine treatment through sensitization of tumor cells to CTL-mediated killing in this model. These findings may have implications for achieving therapeutic control of CTL-resistant tumors in cancer therapy.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Desoxicitidina/análogos & derivados , Receptor ErbB-2/genética , Microambiente Tumoral/efectos de los fármacos , Vacunas de ADN/farmacología , Animales , Anticuerpos/farmacología , Antígeno B7-H1/metabolismo , Desoxicitidina/farmacología , Femenino , Ratones Endogámicos BALB C , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptores de Quimiocina/antagonistas & inhibidores , Linfocitos T Citotóxicos/efectos de los fármacos , Gemcitabina
16.
Oncotarget ; 8(16): 26771-26788, 2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28460461

RESUMEN

In the CT26/HER2 and 4T1.2/HER2 tumor models, CT26/HER2 cells form tumors that continue to grow, whereas 4T1.2/HER2 cells form tumors that eventually regress. Here, we investigated the differences in the behaviors of these two cell lines. When immune cells from 4T1.2/HER2 tumor-bearing animals were stimulated with HER2 class I peptides, they displayed a 2-fold increase in IFN-γ levels, in response to the peptides, HER263-71 and HER2342-350. In contrast, extremely high levels of antigen-non-specific IFN-γ production were observed with immune cells and sera from CT26/HER2 tumor-bearing mice. However, IFN-γ had no effect on tumor progression in the CT26/HER2 model, as determined by an IFN-γ knockout assay. 4T1.2/HER2 tumor-bearing mice displayed CTL activity in response to HER263-71 but not to HER2342-350, whereas no such induction was observed in CT26/HER2 tumor-bearing mice. When 4T1.2/HER2 cell-challenged mice were depleted of CD8+ T cells, they lost their tumor-regressing activity, suggesting an antitumor role of HER263-71-specific CD8+ CTLs in the control of this tumor type. CT26/HER2 cells also expressed CD80. However, CD80-transfected 4T1.2/HER2 and CD80-non-expressing CT26/HER2 cells failed to alter their tumorigenicity, suggesting no role of CD80 in tumor control. Despite increased levels of myeloid-derived suppressor cells in the tumor, they were not associated with tumor progression in the CT26/HER2 model, as determined by a cell depletion assay. Overall, these data show that, contrary to CT26/HER2 tumors, 4T1.2/HER2 tumors regress via the induction of HER263-71-specific CD8+ CTLs and that CD80 is not associated with the regression of these tumors.


Asunto(s)
Neoplasias/inmunología , Neoplasias/metabolismo , Receptor ErbB-2/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Biomarcadores , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Neoplasias/patología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Gemcitabina
17.
Clin Exp Vaccine Res ; 6(2): 135-145, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28775978

RESUMEN

PURPOSE: The goal of this study was to investigate the utility of DNA vaccines encoding Ebola virus glycoprotein (GP) as a vaccine type for the production of GP-specific hybridomas and antibodies. MATERIALS AND METHODS: DNA vaccines were constructed to express Ebola virus GP. Mice were injected with GP DNA vaccines and their splenocytes were used for hybridoma production. Enzyme-linked immunosorbent assays (ELISAs), limiting dilution subcloning, antibody purification methods, and Western blot assays were used to select GP-specific hybridomas and purify monoclonal antibodies (MAbs) from the hybridoma cells. RESULTS: Twelve hybridomas, the cell supernatants of which displayed GP-binding activity, were selected by ELISA. When purified MAbs from 12 hybridomas were tested for their reactivity to GP, 11 MAbs, except for 1 MAb (from the A6-9 hybridoma) displaying an IgG2a type, were identified as IgM isotypes. Those 11 MAbs failed to recognize GP. However, the MAb from A6-9 recognized the mucin-like region of GP and remained reactive to the antigen at the lowest tested concentration (1.95 ng/mL). This result suggests that IgM-secreting hybridomas are predominantly generated by DNA vaccination. However, boosting with GP resulted in greater production of IgG-secreting hybridomas than GP DNA vaccination alone. CONCLUSION: DNA vaccination may preferentially generate IgM-secreting hybridomas, but boosting with the protein antigen can reverse this propensity. Thus, this protein boosting approach may have implications for the production of IgG-specific hybridomas in the context of the DNA vaccination platform. In addition, the purified monoclonal IgG antibodies may be useful as therapeutic antibodies for controlling Ebola virus infection.

18.
DNA Cell Biol ; 25(5): 277-86, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16716117

RESUMEN

We previously reported that E7 subunit and DNA vaccines are both capable of inducing antitumor protection through induction of antigen-specific CTL. In this study, we investigated their ability to control established tumors according to tumor size, vaccine doses, and vaccine delivery routes. Antitumor therapeutic efficacy of both vaccine types was dependent on tumor burden. However, E7 subunit vaccines induced a higher level of antitumor therapeutic activities at the tested dose compared to DNA vaccines. This was concomitant with induction of antibody, CTL, and IFN-gamma responses, as well as histologic changes (heavy infiltration of lymphocytes and presence of apoptotic bodies). In vaccine dose titration assays, 50 and 100 microg of DNA vaccines exhibited an equivalent antitumor efficacy to 0.5 and 1 microg of E7 subunit vaccines, respectively, i.e., a 100-fold difference in E7 dosage, suggesting the importance of vaccine doses for achieving antitumor immunity. Furthermore, tumors of a larger size were controlled by intratumoral injection with E7 subunit vaccines, underscoring the importance of vaccine delivery routes for antitumor therapeutic efficacy. Thus, these data suggest that antitumor therapeutic efficacy of E7 therapeutic vaccines is determined by vaccine doses, vaccine delivery routes, and tumor sizes, and that these vaccines could be another addition to conventional therapy modalities against cervical cancer.


Asunto(s)
Modelos Animales , Neoplasias del Cuello Uterino/terapia , Vacunas de ADN/uso terapéutico , Animales , Secuencia de Bases , Cartilla de ADN , Relación Dosis-Respuesta Inmunológica , Vías de Administración de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Femenino , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Papillomaviridae/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
19.
Cancer Res ; 62(24): 7234-40, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12499264

RESUMEN

An important goal of immunotherapy against human papillomavirus (HPV) infection and the cervical cancer is to control viral infection and the cancer cell growth. Here we investigate the utility of HPV 16 E7 along with CpG-oligodeoxynucleotide (ODN) for protection against HPV-immortalized tumor cells using an animal model. E7+ODN coinjection showed a significant suppression of tumor growth at both prophylactic and therapeutic levels. However, no such effect was observed without addition of both E7 and ODN. We additionally evaluated levels of immune responses by E7+ODN coinjection. E7+ODN resulted in E7-specific antibody (IgG1, IgG2a, IgG2b, and IgG3) and T-helper cell proliferative responses significantly higher than E7 alone. However, CTL responses were induced only by E7+ODN. Moreover, IFN-gamma production was detected only in E7+ODN immunized groups in which IFN-gamma releasing CD4+ (T-helper 1 type) and CD8+ T cells (CTL) were induced only by E7+ODN. Moreover, tumor protection appears to be mediated by CD4+ and in most CD8+ T cells, as determined by in vivo T-cell subset depletion. Taken together, these data suggest that E7+ODN codelivery could be an effective approach to induce E7-specific protective immune responses as a possible immunotherapeutic strategy for cervical cancer.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Pulmonares/inmunología , Oligonucleótidos/inmunología , Proteínas Oncogénicas Virales/inmunología , Proteínas Represoras , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Animales , Especificidad de Anticuerpos , Vacunas contra el Cáncer/farmacología , Línea Celular Transformada , Islas de CpG/inmunología , Femenino , Inmunización , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/clasificación , Inmunoglobulina G/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/virología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos/farmacología , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/farmacología , Papillomaviridae/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/inmunología
20.
Hum Vaccin Immunother ; 11(8): 2012-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25902414

RESUMEN

We previously reported that MC32 cells resist carcinoembryonic antigen (CEA) DNA vaccination by losing their antigen presentation to Ag-specific CTLs in the context of MHC class I antigens in a colon cancer therapeutic model. In this study, we selected 2 tumor cells, MC32-S2-2 and MC32-S4-2, which have the ability to form tumors in CEA DNA vaccine-immunized mice. Wild type MC32 cells grew significantly less in CEA-immunized mice (with Ag-specific CTL lytic activity) than in control mice (with no Ag-specific CTL lytic activity). However, MC32-S2-2 and MC32-S4-2 cells grew at a similar rate in both control and CEA-immunized mice, confirming their resistant status against CEA DNA vaccination. MC32-S2-2 and MC32-S4-2 cells were not susceptible to lysis by CEA-specific CD8+ T cells. Moreover, when MC32-S2-2 and MC32-S4-2 cells were used as stimulating agents of CEA-specific immune cells for IFN-γ production, these cells failed to stimulate the induction of Ag-specific IFN-γ, suggesting a loss of tumor cell recognition by Ag-specific immune cells. However, MC32-S2-2 and MC32-S4-2 cells expressed MHC class I antigens in a manner similar to that of wild type MC32 cells. Finally, Western blot assay confirmed that in MC32-S2-2 and MC32-S4-2 cells, CEA expression remained absent but mouse CEA was expressed. Taken together, these data show that MC32 cells may also be able to achieve resistance to CEA-specific CTLs by antigen loss in this model.


Asunto(s)
Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Neoplasias del Colon/inmunología , Evasión Inmune , Linfocitos T Citotóxicos/inmunología , Animales , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Neoplasias
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda