Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Breast Cancer Res Treat ; 194(1): 65-78, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35482127

RESUMO

PURPOSE: The antigenic targets of immunity and the role of vaccination in breast cancer are unknown. We performed a phase I study of an autologous GM-CSF-secreting breast cancer vaccine in patients with metastatic and stage II-III breast cancer. METHODS: Tumor cells from patients with metastatic (n = 15) and stage II-III (n = 7) disease were transduced with a replication-defective adenoviral vector encoding GM-CSF, and then irradiated. Twelve and seven patients with metastatic and stage II-III disease, respectively, received weekly vaccination for three weeks, followed by every other week until disease progression or vaccine supply was exhausted (metastatic) or until six total vaccine doses were administered (stage II-III). RESULTS: Among those patients with metastatic disease who received vaccinations, eight had progressive disease at two months, three had stable disease for 4-13 months, and one has had no evidence of disease for 13 years. Of the patients with stage II-III disease, five died of metastatic disease between 1.16 and 8.49 years after the start of vaccinations (median 6.24 years) and two are alive as of September 2021. Toxicities included injection site reactions, fatigue, fever, upper respiratory symptoms, joint pain, nausea, and edema. Four of five evaluable patients with metastatic disease developed a skin reaction with immune cell infiltration after the fifth injection of unmodified, irradiated tumor cells. CONCLUSION: We conclude that tumor cells can be harvested from patients with metastatic or stage II-III breast cancer to prepare autologous GM-CSF-secreting vaccines that induce coordinated immune responses with limited toxicity. TRIAL REGISTRATION AND DATE OF REGISTRATION: clinicaltrials.gov, NCT00317603 (April 25, 2006) and NCT00880464 (April 13, 2009).


Assuntos
Neoplasias da Mama , Vacinas Anticâncer , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Vacinas Anticâncer/toxicidade , Estudos de Viabilidade , Feminino , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos
2.
Drug Deliv ; 28(1): 542-549, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33685317

RESUMO

We developed a biocompatible splenic vector for a DNA vaccine against melanoma. The splenic vector is a ternary complex composed of plasmid DNA (pDNA), biodegradable dendrigraft poly-L-lysine (DGL), and γ-polyglutamic acid (γ-PGA), the selective uptake of which by the spleen has already been demonstrated. The ternary complex containing pDNA encoding luciferase (pCMV-Luc) exhibited stronger luciferase activity for RAW264.7 mouse macrophage-like cells than naked pCMV-Luc. Although the ternary complex exhibited strong luciferase activity in the spleen after its tail vein injection, luciferase activity in the liver and spleen was significantly decreased by a pretreatment with clodronate liposomes, which depleted macrophages in the liver and spleen. These results indicate that the ternary complex is mainly transfected in macrophages and is a suitable formulation for DNA vaccination. We applied the ternary complex to a pUb-M melanoma DNA vaccine. The ternary complex containing pUb-M suppressed the growth of melanoma and lung metastasis by B16-F10 mouse melanoma cells. We also examined the acute and liver toxicities of the pUb-M ternary complex at an excess pDNA dose in mice. All mice survived the injection of the excess amount of the ternary complex. Liver toxicity was negligible in mice injected with the excess amount of the ternary complex. In conclusion, we herein confirmed that the ternary complex was mainly transfected into macrophages in the spleen after its tail vein injection. We also showed the prevention of melanoma metastasis by the DNA vaccine and the safety of the ternary complex.


Assuntos
Vacinas Anticâncer/administração & dosagem , Melanoma Experimental/terapia , Transgenes/genética , Vacinas de DNA/administração & dosagem , Animais , Vacinas Anticâncer/toxicidade , Ácido Clodrônico/administração & dosagem , Ácido Clodrônico/farmacologia , Injeções Intravenosas , Lipossomos , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Melanoma Experimental/genética , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Ácido Poliglutâmico/análogos & derivados , Ácido Poliglutâmico/química , Polilisina/química , Células RAW 264.7 , Baço/metabolismo , Transfecção , Vacinas de DNA/toxicidade
3.
Anticancer Res ; 39(4): 1699-1703, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30952708

RESUMO

BACKGROUND/AIM: Previous work in rodent models showed that an autologous tissue vaccine is both a safe and effective approach for treating cancer; however, as a translational step, safety must first be evaluated in a more clinically-relevant model. MATERIALS AND METHODS: An autologous immunotherapy produced from resected tumors, was evaluated in a clinically-relevant canine model to assess safety. Ninety-three dogs with spontaneously occurring tumors received vaccination with inactivated autologous tumor tissue combined with an adjuvant of particulate porcine small intestinal submucosa extracellular matrix (SIS-ECM). Patients were followed to assess the occurrence of adverse events, overall survival, and tumor recurrence and/or metastasis. RESULTS: A small number (12%) of patients experienced limited, mild pyrexia, injection site swelling, or lethargy, all resolving without clinical intervention. CONCLUSION: Autologous whole cell cancer immunotherapy can be used safely in the canine model of cancer and represents a safe approach for the treatment for cancer.


Assuntos
Antígenos de Neoplasias/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Doenças do Cão/terapia , Imunoterapia/veterinária , Neoplasias/veterinária , Adjuvantes Imunológicos , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/toxicidade , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Doenças do Cão/diagnóstico , Doenças do Cão/imunologia , Doenças do Cão/cirurgia , Cães , Feminino , Imunoterapia/efeitos adversos , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Masculino , Margens de Excisão , Neoplasia Residual , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/terapia , Sus scrofa
4.
Int Immunopharmacol ; 48: 55-60, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28463787

RESUMO

CIGB 247 is a novel cancer therapeutic vaccine based on human vascular endothelial growth factor (VEGF) variant molecule as antigen, in combination with a bacterial adjuvant. This vaccine candidate has previously demonstrated efficacy and safety in mice, rats, rabbits and non-human primates. In the present study we evaluated the effects on the clinical, hematological and biochemical parameters of CIGB 247 vaccine in Chlorocebus aethiops monkeys. Three groups of monkeys were immunized with three doses of vaccine formulation to measure physiological values of clinical, hematological and serum biochemical parameters. Monkeys' body weight and temperature were kept stable and close to standard values throughout the study. Variations in the levels of red blood cells and hemoglobin were observed among the different groups for all injected doses, but these hematological parameters recovered normal values at the end of the study. On the other hand, biochemical parameters such as the total bilirubin and total protein counts showed variations along the study, while they were not associated with the test substance. In summary, no negative effects on clinical, hematological and biochemical parameters were detected. Together, our results put forward the potential and support the safety of the CIGB 247 vaccine candidate for use in clinical applications. The data presented here can be used to estimate a human dosing regimen from preclinical data.


Assuntos
Vacinas Anticâncer/toxicidade , Animais , Chlorocebus aethiops , Eritrócitos/efeitos dos fármacos , Hemoglobinas/metabolismo , Masculino , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
5.
Oncotarget ; 5(24): 12803-10, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25296974

RESUMO

Our previous data demonstrated profound anti-tumor and anti-metastatic effects of p62 (sqstm1) DNA vaccine in rodents with various types of transplantable tumors. Testing anti-cancer medicine in dogs as an intermediary step of translational research program provides two major benefits. First, clinical data collected in target animals is required for FDA/USDA approval as a veterinary anti-cancer drug or vaccine. It is noteworthy that the veterinary community is in need of novel medicine for the prevention and treatment of canine and feline cancers. The second more important benefit of testing anti-cancer vaccines in dogs is that spontaneous tumors in dogs may provide invaluable information for human trials. Here, we evaluated the effect(s) of p62 DNA vaccine on mammary tumors of dogs. We found that p62 DNA vaccine administered i.m. decreased or stabilized growth of locally advanced lesions in absence of its overall toxic effects. The observed antitumor activity was associated with lymphocyte infiltration and tumor encapsulation via fibrotic reaction. This data justifies both human clinical trials and veterinary application of p62 DNA vaccine.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/toxicidade , Doenças do Cão/terapia , Neoplasias Mamárias Animais/terapia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/toxicidade , Animais , Vacinas Anticâncer/imunologia , Cães , Feminino , Humanos , Masculino , Projetos Piloto , Proteína Sequestossoma-1 , Testes de Toxicidade , Vacinas de DNA/imunologia
6.
Sci Rep ; 3: 2203, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23857555

RESUMO

Zn-, and Mg-containing tricalcium phosphates (TCPs) loaded with a hydrothermal extract of a human tubercle bacillus (HTB) were prepared by immersing Zn-TCP and Mg-TCP in HTB-containing supersaturated calcium phosphate solutions. The in vitro and in vivo immunogenic activities of the HTB-loaded Zn-, and Mg-TCPs (Zn-Ap-HTB and Mg-Ap-HTB, respectively) were evaluated as potential immunopotentiating adjuvants for cancer immunotherapy. The Zn-Ap-HTB and Mg-Ap-HTB adjuvants showed no obvious cytotoxicity and more effectively stimulated granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion by macrophage-like cells than unprocessed HTB or HTB-loaded TCP (T-Ap-HTB) in vitro. Zn-Ap-HTB and Mg-Ap-HTB mixed with liquid-nitrogen-treated tumor tissue markedly inhibited the in vivo development of rechallenged Lewis lung carcinoma (LLC) cells compared with T-Ap-HTB and the unprocessed HTB mixed liquid-nitrogen-treated tumor tissue. Zn-Ap-HTB and Mg-Ap-HTB contributed to eliciting potent systemic antitumor immunity in vivo.


Assuntos
Adjuvantes Imunológicos , Fosfatos de Cálcio/química , Fosfatos de Cálcio/imunologia , Vacinas Anticâncer , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Bacillus/química , Bacillus/imunologia , Fosfatos de Cálcio/administração & dosagem , Fosfatos de Cálcio/toxicidade , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Magnésio/química , Camundongos , Neoplasias/mortalidade , Baço/imunologia , Baço/metabolismo , Vacinas Conjugadas , Zinco/química
7.
Mol Ther ; 21(5): 1076-86, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23531551

RESUMO

The industrial development of active immunotherapy based on live-attenuated bacterial vectors has matured. We developed a microsyringe for antigen delivery based on the type III secretion system (T3SS) of P. aeruginosa. We applied the "killed but metabolically active" (KBMA) attenuation strategy to make this bacterial vector suitable for human use. We demonstrate that attenuated P. aeruginosa has the potential to deliver antigens to human antigen-presenting cells in vitro via T3SS with considerable attenuated cytotoxicity as compared with the wild-type vector. In a mouse model of cancer, we demonstrate that this KBMA strain, which cannot replicate in its host, efficiently disseminates into lymphoid organs and delivers its heterologous antigen. The attenuated strain effectively induces a cellular immune response to the cancerous cells while lowering the systemic inflammatory response. Hence, a KBMA P. aeruginosa microsyringe is an efficient and safe tool for in vivo antigen delivery.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/imunologia , Imunoterapia , Pseudomonas aeruginosa/imunologia , Animais , Sistemas de Secreção Bacterianos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Furocumarinas/farmacologia , Humanos , Imunidade Celular , Tecido Linfoide/imunologia , Tecido Linfoide/microbiologia , Camundongos , Mutação , Neoplasias/imunologia , Neoplasias/prevenção & controle , Neoplasias/terapia , Fármacos Fotossensibilizantes/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Regul Toxicol Pharmacol ; 64(3): 425-34, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23085371

RESUMO

Our goal was to assess the toxicity of two strengths (200 and 400 µg) of HER1 cancer vaccine (Center of Molecular Immunology, Cuba), presented in two different formulations, in Sprague Dawley rats after repeated intramuscular administration (14 days). Four groups (5 animals/sex) were established: Control, Placebo (adjuvant), and two Treated groups receiving a dose representing ten times of human total dose (10×), 28.6 and 57.1 µg/kg. Clinical observations, body weight and rectal temperature were measured during the study. Clinical pathology analysis was performed, besides gross necropsy and histological examination of tissues on animals at the end of the assay. The assay ended with a 100% survival. Injection site damage, with the presence of cysts and granulomas, was observed in adjuvant and vaccine treated groups, with most severe cases predominating at higher strength. Administration of Placebo and Her1 vaccine induced increase in polymorphonuclear cells, with relative lymphopenia conditioned by primary neutrophilia. In summary, results suggest that Her1 immunization was capable of inducing an inflammatory effect at the injection site, leading to systemic alterations, more significant at higher strength (400 µg, 57.1 µg/kg), probably affected by the immunizations' schedule used. The vaccine was shown to be well tolerated without any obvious signs of systemic toxicity, with findings largely attributable to the adjuvant used.


Assuntos
Vacinas Anticâncer/toxicidade , Fator de Crescimento Epidérmico/imunologia , Inflamação/induzido quimicamente , Animais , Vacinas Anticâncer/administração & dosagem , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Injeções Intramusculares , Masculino , Neutrófilos/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Cell Immunol ; 270(2): 135-44, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21741036

RESUMO

We had previously shown that activated NKT cells licensed B cells to be immunogenic antigen-presenting cells and helped to elicit a wide spectrum of cancer targeted immune responses. In the current study, we sought to verify the safety of αGalCer-loaded, and adenovirus-transduced B cell-based vaccines, together with mechanism of action. Intravenously injected αGalCer-loaded, antigen-expressing B cells rapidly localized in the spleen and directly primed CD8(+) T cells in an antigen-specific manner. The transferred antigen was sustained for at least 30 days. While some injected B cells produced nonspecific IgG, the antigen-specific IgG response was completely dependent on endogenous B cells. The liver was one of the main tissues where injected B cells were retained; however, we could not find the signs of liver toxicity. Our results demonstrate that αGalCer-loaded, antigen-expressing B cells behave as "antigen-presenting" cells that stimulate endogenous antigen-specific T cells and B cells in vivo without significant toxicity.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Células T Matadoras Naturais/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Especificidade de Anticorpos , Linfócitos B/transplante , Vacinas Anticâncer/toxicidade , Galactosilceramidas/administração & dosagem , Galactosilceramidas/imunologia , Imunoglobulina G/biossíntese , Imunoterapia Ativa , Ligantes , Fígado/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/imunologia , Neoplasias/terapia , Baço/imunologia , Transfecção
10.
Mol Med Rep ; 4(5): 857-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21720718

RESUMO

Tumor growth is partly dependent on tumor-associated angiogenesis, which is regulated by angiogenic growth factors. As the first angiogenic growth factor to be identified, basic fibroblast growth factor (bFGF) plays a major role in angiogensis and tumor growth and has been an effective target for anti-tumor therapy. However, due to its low immunogenicity, injection with bFGF alone cannot stimulate the body to produce a strong immune response. In this study, we investigated the role of CF (containing bFGF and CRM197) assisted by CpG and alum in enhancing antigen-specific immune response and suppressing the growth of murine colon carcinoma. The results revealed that compared to bFGF, CF could not stimulate NIH-3T3 fibroblast proliferation even at a concentration of 10 µg/ml in vitro. In vivo, the CF-CpG-alum produced a stronger antigen-specific immune response and inhibited tumor growth. The anti-tumor activity was associated with generating antigen-specific antibody, suppressing angiogenesis, promoting the apoptosis of tumor cells and inducing the mixed Th1 and Th2 responses. This indicates that CRM197 may be an innovative intramolecular adjuvant and provides a rational preservation for mouse CT26 colon carcinoma.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas de Transporte/imunologia , Fator 2 de Crescimento de Fibroblastos/imunologia , Imunidade/imunologia , Vacinas Conjugadas/imunologia , Compostos de Alúmen , Animais , Apoptose , Proteínas de Bactérias/toxicidade , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Feminino , Vetores Genéticos/genética , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Neovascularização Patológica/patologia , Oligodesoxirribonucleotídeos/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/isolamento & purificação , Resultado do Tratamento , Vacinas Conjugadas/toxicidade
11.
Clin Cancer Res ; 17(14): 4844-53, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21632857

RESUMO

PURPOSE: The use of tumor-derived proteins as cancer vaccines is complicated by tolerance to these self-antigens. Tolerance may be broken by immunization with activated, autologous, ex vivo generated and antigen-loaded, antigen-presenting cells (APC); however, targeting tumor antigen directly to APC in vivo would be a less complicated strategy. We wished to test whether targeted delivery of an otherwise poorly immunogenic, soluble antigen to APC through their mannose receptors (MR) would induce clinically relevant immunity. EXPERIMENTAL DESIGN: Two phase I studies were conducted with CDX-1307, a vaccine composed of human chorionic gonadotropin beta-chain (hCG-ß) fused to an MR-specific monoclonal antibody, administered either locally (intradermally) or systemically (intravenously) in patients with advanced epithelial malignancies. An initial dose escalation of single-agent CDX-1307 was followed by additional cohorts of CDX-1307 combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) and the Toll-like receptor (TLR) 3 agonist polyinosinic-polycytidylic acid (poly-ICLC) and TLR7/8 agonist resiquimod to activate the APC. RESULTS: CDX-1307 induced consistent humoral and T-cell responses to hCG-ß when coadministered with TLR agonists. Greater immune responses and clinical benefit, including the longest duration of stable disease, were observed with immunization combined with local TLR agonists. Immune responses were induced equally efficiently in patients with elevated and nonelevated levels of serum hCG-ß. Antibodies within the serum of vaccinated participants had tumor suppressive function in vitro. Toxicity consisted chiefly of mild injection site reactions. CONCLUSIONS: APC targeting and activation induce adaptive immunity against poorly immunogenic self-antigens which has implications for enhancing the efficacy of cancer immunotherapy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Autoantígenos/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias/imunologia , Neoplasias/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Receptores Toll-Like/agonistas , Células Apresentadoras de Antígenos/metabolismo , Autoantígenos/metabolismo , Vacinas Anticâncer/farmacocinética , Vacinas Anticâncer/toxicidade , Linhagem Celular Tumoral , Gonadotropina Coriônica Humana Subunidade beta/sangue , Gonadotropina Coriônica Humana Subunidade beta/imunologia , Feminino , Humanos , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Masculino , Estadiamento de Neoplasias , Neoplasias/patologia , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/toxicidade , Pele/imunologia , Pele/metabolismo , Pele/patologia , Receptores Toll-Like/metabolismo , Resultado do Tratamento
12.
J BUON ; 14 Suppl 1: S165-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19785060

RESUMO

Producing effective therapeutic vaccines has proved much more difficult and challenging than developing cancer preventive vaccines. Despite huge research in the area of cancer immunology, FDA/EMEA have not approved any type of cancer treatment vaccine so far. More than 99% of cervical cancers have detectable amounts of human papillomavirus (HPV) DNA. Integration of high-risk HPV into the host cell genome is followed by continual expression of HPV E6 and E7 oncoproteins, making them excellent targets for developing vaccines which could be used in high grade precancerous (CIN) lesions or invasive cancer or in the prevention of cancer recurrence. Therapeutic cervical cancer vaccines have been extensively studied. Strategies used were vaccination with HPV peptides or proteins, alone or in pulsed dendritic cells, DNA vaccines, virus-like particles or viral and bacterial vectors. Lovaxin-C is a recombinant live-attenuated Listeria monocytogenes (Lm) that secretes the antigen HPV-16 E7 fused to a non-hemolytic listeriolysin O protein. In a phase I study Lovaxin-C was administered to advanced cervical cancer patients refractory to existing therapies. The dose-limiting toxicity was hypotension and flue-like syndrome. There were no serious adverse events. Specific T-cell response was detected as well as clinical response to Lovaxin-C. Several other therapeutic HPV vaccines are in clinical development and in most of the studies specific immunological and clinical responses were seen. Efficacious therapeutic vaccine for the treatment of cervical cancer should be expected in the near future.


Assuntos
Vacinas Anticâncer/toxicidade , Neoplasias do Colo do Útero/imunologia , Ampicilina/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Relação Dose-Resposta a Droga , Estudos de Viabilidade , Feminino , Humanos , Hipotensão/induzido quimicamente , Segurança , Neoplasias do Colo do Útero/prevenção & controle
13.
Cancer Immunol Immunother ; 58(2): 201-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18523770

RESUMO

Previous studies have shown that there are profuse lymphatic tissues under the intestinal mucous membrane. Moreover, vaccine administered orally can elicit both mucous membrane and system immune response simultaneously, accordingly induce tumor-specific cytotoxic T lymphocyte. As a result, the oral route is constituted the preferred immune route for vaccine delivery theoretically. However, numerous vaccines especially protein/peptide vaccines remain poorly available when administered by this route. Nanoemulsion has been shown as a useful vehicle can be developed to enhance the antitumor immune response against antigens encapsulated in it and it is good for the different administration routes. Of particular interest is whether the protein vaccine following peroral route using nanoemulsion as delivery carrier can induce the same, so much as stronger antitumor immune response to following conventional ways such as subcutaneous (sc.) or not. Hence, in the present study, we encapsulated the MAGE1-HSP70 and SEA complex protein in nanoemulsion as nanovaccine NE (MHS) using magnetic ultrasound method. We then immuned C57BL/6 mice with NE (MHS), MHS alone or NE (-) via po. or sc. route and detected the cellular immunocompetence by using ELISpot assay and LDH release assay. The therapeutic and tumor challenge assay were examined then. The results showed that compared with vaccination with MHS or NE (-), the cellular immune responses against MAGE-1 could be elicited fiercely by vaccination with NE (MHS) nanoemulsion. Furthermore, encapsulating MHS in nanoemulsion could delay tumor growth and defer tumor occurrence of mice challenged with B16-MAGE-1 tumor cells. Especially, the peroral administration of NE (MHS) could induce approximately similar antitumor immune responses to the sc. administration, but the MHS unencapsulated with nanoemulsion via po. could induce significantly weaker antitumor immune responses than that via sc., suggesting nanoemulsion as a promising carrier can exert potent antitumor immunity against antigen encapsulated in it and make the tumor protein vaccine immunizing via po. route feasible and effective. It may have a broad application in tumor protein vaccine.


Assuntos
Antígenos de Neoplasias/farmacologia , Vacinas Anticâncer/farmacologia , Proteínas de Choque Térmico HSP70/farmacologia , Nanopartículas , Proteínas de Neoplasias/farmacologia , Administração Oral , Animais , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Linhagem Celular Tumoral , Vias de Administração de Medicamentos , Emulsões/administração & dosagem , Emulsões/farmacologia , Proteínas de Choque Térmico HSP70/administração & dosagem , Proteínas de Choque Térmico HSP70/imunologia , Ativação Linfocitária , Antígenos Específicos de Melanoma , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/ultraestrutura , Proteínas de Neoplasias/administração & dosagem , Proteínas de Neoplasias/imunologia , Linfócitos T/imunologia
14.
J Clin Oncol ; 26(30): 4973-80, 2008 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-18809608

RESUMO

PURPOSE: A phase I/II trial was performed to evaluate the safety and immunogenicity of a novel melanoma vaccine comprising six melanoma-associated peptides defined as antigenic targets for melanoma-reactive helper T cells. Source proteins for these peptides include MAGE proteins, MART-1/MelanA, gp100, and tyrosinase. PATIENTS AND METHODS: Thirty-nine patients with stage IIIB to IV melanoma were vaccinated with this six-peptide mixture weekly at three dose levels, with a preceding phase I dose escalation and subsequent random assignment among the dose levels. Helper T-lymphocyte responses were assessed by in vitro proliferation assay and delayed-type hypersensitivity skin testing. Patients with measurable disease were evaluated for objective clinical response by Response Evaluation Criteria in Solid Tumors. RESULTS: Vaccination with the helper peptide vaccine was well tolerated. Proliferation assays revealed induction of T-cell responses to the melanoma helper peptides in 81% of patients. Among 17 patients with measurable disease, objective clinical responses were observed in two patients (12%), with response durations of 1 and 3.9+ years. Durable stable disease was observed in two additional patients for periods of 1.8 and 4.6+ years. CONCLUSION: Results of this study support the safety and immunogenicity of a vaccine comprised of six melanoma helper peptides. There is also early evidence of clinical activity.


Assuntos
Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Melanócitos/imunologia , Melanoma/imunologia , Melanoma/prevenção & controle , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/prevenção & controle , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Diferenciação/imunologia , Antígenos de Neoplasias/imunologia , Proliferação de Células , Feminino , Humanos , Antígeno MART-1 , Masculino , Melanoma/patologia , Glicoproteínas de Membrana/imunologia , Pessoa de Meia-Idade , Monofenol Mono-Oxigenase/imunologia , Proteínas de Neoplasias/imunologia , Estadiamento de Neoplasias , Neoplasias Cutâneas/patologia , Testes Cutâneos , Linfócitos T Auxiliares-Indutores/imunologia , Vitiligo/induzido quimicamente , Antígeno gp100 de Melanoma
15.
J Immunother ; 31(6): 545-54, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18528300

RESUMO

Owing to the absence of alphaGal epitopes in human cells and constant stimulation of the immune system by the symbiotic bacterial flora, humans develop high titers of natural antibodies against these epitopes. It has been demonstrated that syngeneic whole cell vaccines modified to express alphaGal epitopes could be used to generate a potent anticancer vaccine. In this study, we tested whether allogeneic whole cell cancer vaccines modified to express alphaGal epitopes would be effective for the treatment of murine melanoma. The alpha(1,3)galactosyltransferase (alphaGT) knockout mice (H-2) with preexisting subcutaneous and pulmonary tumors [alphaGal B16, H-2] received therapeutic vaccinations with S91M3alphaGal (H-2) whole cell allogeneic vaccines. These mice had better survival and reduced pulmonary metastasis burden compared with control mice treated with S91M3 vaccine cells. Vaccination with S91M3alphaGal-induced cytotoxic CD8 T cells recognizing the syngeneic alphaGal B16 tumors measured by adoptive transfer to recipients bearing pulmonary metastases. The presence of allo-antigens did not dominate the induction of immunity to "cryptic" tumor antigens and had helped in the generation of a more efficient vaccine to treat preexisting tumors when compared with classic autologous vaccines. Vaccination with allogeneic alphaGal vaccines did not induce signs of toxicity including changes in weight, hematology, chemistry, and histopathology of major perfused organs or autoimmunity in long-term murine models for breast, lung, and melanoma. This study established the safety and efficacy data of allogeneic alphaGal whole cell vaccines and constituted the basis for the initiation of human clinical trials to treat human malignancies.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia Adotiva , Melanoma Experimental/imunologia , Neoplasias Experimentais/terapia , Trissacarídeos/genética , Animais , Vacinas Anticâncer/toxicidade , Linhagem Celular Tumoral , Feminino , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias Experimentais/imunologia , Neoplasias Cutâneas/terapia
16.
Hautarzt ; 59(6): 475-83, 2008 Jun.
Artigo em Alemão | MEDLINE | ID: mdl-18449519

RESUMO

To treat malignant melanoma successfully currently means to recognize the tumor at an early stage and to remove it immediately. Aside from individual cases, available treatment modalities are not able to increase survival, especially in the palliative situation. Thus innovative experimental approaches are urgently needed to strongly improve the palliative and adjuvant treatment of melanoma. Anti-tumor effects are expected from targeted therapies, which are directed against defined molecules decisive for tumor pathogenesis. Crucial points of attack are signaling pathways, angiogenesis and apoptosis resistance. New diagnostic and therapeutic developments have enhanced the efficacy of chemotherapies. Increasing insights into tumor immunology provide new treatment approaches of vaccination, cell transfer and especially of blocking immune tolerance mechanisms. It will be challenging for the future to identify and characterize more precisely those patients who might most benefit from a certain treatment approach.


Assuntos
Antineoplásicos/uso terapêutico , Drogas em Investigação/uso terapêutico , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Transferência Adotiva , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/toxicidade , Antígenos CD/genética , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Apoptose/genética , Antígeno CTLA-4 , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/toxicidade , Análise Mutacional de DNA , Drogas em Investigação/toxicidade , Humanos , Melanoma/genética , Melanoma/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Qualidade de Vida , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
17.
Blood ; 111(6): 3116-25, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18202224

RESUMO

Identification of Toll-like receptors (TLRs) and their ligands, and tumor necrosis factor-tumor necrosis factor receptor (TNF-TNFR) pairs have provided the first logical, hypothesis-based strategies to molecularly concoct adjuvants to elicit potent cell-mediated immunity via activation of innate and adaptive immunity. However, isolated activation of one immune pathway in the absence of others can be toxic, ineffective, and detrimental to long-term, protective immunity. Effective engineered vaccines must include agents that trigger multiple immunologic pathways. Here, we report that combinatorial use of CD40 and TLR agonists as a cancer vaccine, compared with monotherapy, elicits high frequencies of self-reactive CD8(+) T cells, potent tumor-specific CD8(+) memory, CD8(+) T cells that efficiently infiltrate the tumor-burdened target organ; therapeutic efficacy; heightened ratios of CD8(+) T cells to FoxP3(+) cells at the tumor site; and reduced hepatotoxicity. These findings provide intelligent strategies for the formulation of multifactorial vaccines to achieve maximal efficacy in cancer vaccine trials in humans.


Assuntos
Adjuvantes Imunológicos/toxicidade , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Animais , Anticorpos/imunologia , Antígenos CD40/agonistas , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/metabolismo , Linhagem Celular Tumoral , Transplante de Células , Memória Imunológica/imunologia , Imunoterapia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/lesões , Pulmão/imunologia , Pulmão/cirurgia , Masculino , Melanoma/imunologia , Melanoma/patologia , Melanoma/terapia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo
18.
J Transl Med ; 6: 6, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18221542

RESUMO

BACKGROUND: Sixteen melanoma patients (1 stage IIC, 8 stage III, and 7 stage IV) were treated in a Phase I study with a vaccine (DC/Apo-Nec) composed of autologous dendritic cells (DCs) loaded with a mixture of apoptotic/necrotic allogeneic melanoma cell lines (Apo-Nec), to evaluate toxicity and immune responses. Also, IL-10 1082 genotype was analyzed in an effort to predict disease progression. METHODS: PBMC were obtained after leukapheresis and DCs were generated from monocytes cultured in the presence of GM-CSF and IL-4 in serum-free medium. Immature DCs were loaded with gamma-irradiated Apo-Nec cells and injected id without adjuvant. Cohorts of four patients were given four vaccines each with 5, 10, 15, or 20 x 106 DC/Apo-Nec cell per vaccine, two weeks apart. Immune responses were measured by ELISpot and tetramer analysis. Il-10 genotype was measured by PCR and corroborated by IL-10 production by stimulated PBMC. RESULTS: Immature DCs efficiently phagocytosed melanoma Apo-Nec cells and matured after phagocytosis as evidenced by increased expression of CD83, CD80, CD86, HLA class I and II, and 75.2 +/- 16% reduction in Dextran-FITC endocytosis. CCR7 was also up-regulated upon Apo-Nec uptake in DCs from all patients, and accordingly DC/Apo-Nec cells were able to migrate in vitro toward MIP-3 beta. The vaccine was well tolerated in all patients. The DTH score increased significantly in all patients after the first vaccination (Mann-Whitney Test, p < 0.05). The presence of CD8+T lymphocytes specific to gp100 and Melan A/MART-1 Ags was determined by ELISpot and tetramer analysis in five HLA-A*0201 patients before and after vaccination; one patient had stable elevated levels before and after vaccination; two increased their CD8 + levels, one had stable moderate and one had negligible levels. The analysis of IL-10 promoter -1082 polymorphism in the sixteen patients showed a positive correlation between AA genotype, accompanied by lower in vitro IL-10 production by stimulated PBMC, and faster melanoma progression after lymph nodes surgery (p = 0.04). With a mean follow-up of 49.5 months post-surgery, one stage IIC patient and 7/8 stage III patients remain NED but 7/7 stage IV patients have progressed. CONCLUSION: We conclude that DC/Apo-Nec vaccine is safe, well tolerated and it may induce specific immunity against melanoma Ags. Patients with a low-producing IL-10 polymorphism appear to have a worst prognosis. TRIAL REGISTRATION: Clinicaltrials.gov (NHI) NCT00515983.


Assuntos
Vacinas Anticâncer/toxicidade , Células Dendríticas/imunologia , Células Dendríticas/transplante , Interleucina-10/genética , Melanoma/imunologia , Melanoma/patologia , Adolescente , Adulto , Apoptose , Progressão da Doença , Feminino , Genótipo , Humanos , Hipersensibilidade Tardia , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Necrose , Estadiamento de Neoplasias , Seleção de Pacientes , Fagocitose , Polimorfismo Genético , Valor Preditivo dos Testes , Regiões Promotoras Genéticas , Transplante Autólogo
19.
Cancer Sci ; 98(12): 1965-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17919310

RESUMO

The aim of this clinical trial was to investigate the toxicity and immunological responses of personalized peptide vaccination for cytokine-refractory metastatic renal cell carcinoma patients. Patients were confirmed to be human leukocyte antigen (HLA)-A24 or HLA-A2 positive and had histologically confirmed renal cell carcinoma. Ten patients were enrolled in the present study. The peptides to be administered were determined based on the presence of peptide-specific cytotoxic T lymphocyte precursors in peripheral blood mononuclear cells (PBMC) and peptide-specific IgG in the plasma of cancer patients. Patients received subcutaneous injections of four different peptides (3 mg/peptide) every 2 weeks. Vaccinations were well tolerated without any major adverse events. A minimal increase in peptide-specific interferon-gamma production in postvaccination PBMC was observed, regardless of higher levels of cytotoxic T lymphocyte activity in prevaccination PBMC. In contrast, an increase in peptide-specific IgG levels of postvaccination (sixth) plasma was observed in the majority of patients. After progression, five patients received interleukin-2 therapy and continuous vaccination, with survival of 31, 25, 23, 17, and 15 months, but interleukin-2 did not impede humoral responses boosted by the vaccination. These results encourage further clinical trials of personalized peptide vaccinations.


Assuntos
Vacinas Anticâncer/toxicidade , Carcinoma de Células Renais/imunologia , Neoplasias Renais/imunologia , Vacinas de Subunidades Antigênicas/toxicidade , Carcinoma de Células Renais/patologia , Terapia Combinada , Citocinas/uso terapêutico , Antígenos HLA-A/imunologia , Antígeno HLA-A2/imunologia , Antígeno HLA-A24 , Humanos , Imunoglobulina G/sangue , Neoplasias Renais/patologia , Metástase Neoplásica , Estadiamento de Neoplasias , Seleção de Pacientes
20.
Cancer Biother Radiopharm ; 22(3): 309-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17651037

RESUMO

Seventy-four (74) patients with metastatic melanoma were treated with patient-specific vaccines derived from autologous tumor cell lines. Cryopreserved irradiated tumor cells were injected weekly for 3 weeks, then monthly for 5 months. At a median follow up >6 years, the median event-free survival (EFS) was 4.5 months, with 13 patients alive and progression free 6-12 years later. Median overall survival (OS) was 20.5 months, with 29% 5-year OS. Tumor response rate was 9% among the 35 patients with evaluable disease who received at least 3 injections. Better survival was observed for patients who had minimal rather than clinically evident metastatic disease at the time vaccine therapy was initiated (5-yr OS 47% vs. 13%; p < 0.0001), received granulocyte-macrophage colony-stimulating factor and/or interferon gamma as an adjuvant (5-yr EFS 26% vs. 0%; p < 0.0001) or received an average of <7 million cells for each of the first 3 injections, compared to those who received 7-11.9 million or >12 million cells per injection (5-yr EFS OS 35% vs. 24%; p = 0.041 and p = 0.034). There was a trend toward better EFS for those who had a positive delayed type hypersensitivity (DTH) reaction to an intradermal injection of 1 million irradiated tumor cells at baseline, or converted to positive after 3 injections, compared to those whose DTH remained negative (5-yr EFS 39% vs. 18%; p = 0.159). This treatment approach is feasible, produces minimal toxicity, and is associated with longterm survival in a significant proportion of patients.


Assuntos
Vacinas Anticâncer/toxicidade , Vacinas Anticâncer/uso terapêutico , Melanoma/imunologia , Linhagem Celular Tumoral , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Humanos , Imunoterapia/métodos , Espectroscopia de Ressonância Magnética , Melanoma/diagnóstico por imagem , Melanoma/mortalidade , Melanoma/patologia , Metástase Neoplásica/imunologia , Seleção de Pacientes , Estudos Retrospectivos , Análise de Sobrevida , Sobreviventes , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...