Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 421
Filtrar
1.
Prostate Cancer Prostatic Dis ; 15(3): 289-95, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22391584

RESUMO

BACKGROUND: Vaccine therapy in combination with radiation therapy may improve distant and/or local control in prostate cancer. We present long-term follow-up data on the secondary and exploratory endpoints of safety and biochemical failure, respectively, from patients with clinically localized prostate cancer treated definitively with a poxviral vector-based therapeutic vaccine combined with external beam radiation therapy (EBRT). METHODS: Thirty-six prostate cancer patients received definitive EBRT plus vaccine. A total of 18 patients were treated with adjuvant standard-dose interleukin-2 (S-IL-2) (4 MIU m(-2)) and 18 were treated with very low-dose IL-2 (M-IL-2) (0.6 MIU m(-2)). Seven patients were treated with EBRT alone. Twenty-six patients treated with EBRT plus vaccine returned for follow-up, and we reviewed the most recent labs and clinical notes of the remaining patients. RESULTS: Median follow-up for the S-IL-2, M-IL-2 and EBRT-alone groups was 98, 76 and 79 months, respectively. Actuarial 5-year PSA failure-free probability was 78%, 82% and 86% (P=0.58 overall), respectively. There were no significant differences between the actuarial overall survival and the prostate cancer-specific survival between the two vaccine arms. Of the 26 patients who returned for follow-up, Radiation Therapy Oncology Group grade ≥2 genitourinary (GU) and gastrointestinal (GI) toxicity was seen in 19% and 8%, respectively, with no difference between the arms (P=1.00 and P=0.48 for grade ≥2 GU and GI toxicity, respectively). In all, 12 patients were evaluated for PSA-specific immune responses, and 1 demonstrated a response 66 months post-enrollment. CONCLUSIONS: We demonstrate that vaccine combined with EBRT does not appear to have significant differences with regard to PSA control or late-term toxicity compared with standard treatment. We also found limited evidence of long-term immune response following vaccine therapy.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias da Próstata/terapia , Idoso , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Seguimentos , Humanos , Interleucina-2/administração & dosagem , Interleucina-2/efeitos adversos , Interleucina-2/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/radioterapia , Resultado do Tratamento
2.
Curr Oncol ; 18(3): e150-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21655153

RESUMO

With the recent approval by the U.S. Food and Drug Administration of the first therapeutic vaccine for cancer, the long-awaited goal of harnessing a patient's immune system to attack cancer through this modality is finally realized. However, as researchers in the field of cancer immunotherapy continue to perform randomized definitive studies, much remains to be learned about potential surrogate endpoints and appropriate patient populations for therapeutic vaccines. The present review addresses available data from clinical trials of immunotherapeutic agents relevant to the selection of appropriate patient populations. We believe that the weight of evidence supports the use of immunotherapy earlier in the disease course and in patients with less aggressive disease, and that the relevant findings have important implications for the design of clinical trials with therapeutic vaccines.

3.
Curr Oncol ; 14(6): 238-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18080016

RESUMO

Vaccines constitute a potential new therapeutic approach for a range of human cancers. Unlike other therapeutics, vaccines initiate a dynamic process in the host immune system that can be exploited with subsequent therapies. Indeed, recent preclinical and clinical studies with cancer vaccines have provided evidence that this unique therapeutic modality should lead to consideration of new paradigms in both clinical trial design and endpoints and in combination therapies. The present article reviews and sets out a rationale for these new paradigms, with a focus on prostate cancer.

4.
Vaccine ; 23(22): 2909-21, 2005 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-15780740

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a potent immune stimulant when administered with different vaccines. Optimal use of GM-CSF resides in its ability to act locally to stimulate the proliferation and maturation of professional antigen-presenting cells (APCs) (i.e., Langerhans' cells) at the injection site. GM-CSF was engineered into a replication-incompetent recombinant avian (fowlpox) virus (rF-GM-CSF) and a single subcutaneous injection resulted in a sustained enrichment of activated dendritic cells within the regional draining lymph nodes. Those changes were attributed to local GM-CSF production at the injection site by rF-GM-CSF-infected cells. Studies were carried out in which mice were administered different types of beta-galactosidase (beta-gal)-based vaccines--whole protein, peptide, recombinant poxviruses--and GM-CSF was administered either as a single injection of rF-GM-CSF or four daily bolus injections of the recombinant protein. The use of rF-GM-CSF either improved the immune adjuvant effect, as observed for poxvirus-based vaccines, or was equivalent to rGM-CSF, as observed with the beta-gal protein vaccine. It is important to note that with either the replication-competent (vaccinia) or replication-incompetent (fowlpox) vaccines expressing LacZ, strong CTL responses directed against beta-gal were induced only when rF-GM-CSF was used as the immune adjuvant. Engineering GM-CSF into a recombinant fowlpox virus offers an excellent vehicle for the delivery of this cytokine as an immune adjuvant with specific vaccine platforms. In particular, delivery of GM-CSF via the rF-GM-CSF construct would be preferred over bolus injections of rGM-CSF when used as an immune adjuvant with whole protein or recombinant poxvirus-based vaccines. The study underscores the importance of defining the appropriate delivery form of an immune adjuvant, such as GM-CSF, relative to the immunization strategy to maximize the host immune responses against a specific antigen.


Assuntos
Avipoxvirus/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Animais , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Linfócitos T Citotóxicos/imunologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-12751840

RESUMO

The activation of a T cell has been shown to require two signals via molecules present on professional antigen presenting cells: signal 1, via a peptide/MHC complex, and signal 2, via a costimulatory molecule. Here, the role of three costimulatory molecules in the activation of T cells was examined. Poxvirus (vaccinia and avipox) vectors were employed because of their ability to efficiently express multiple genes. Murine cells provided with signal 1 and infected with either recombinant vaccinia or avipox vectors containing a TRIad of COstimulatory Molecules (B7-1/ICAM-1/LFA-3, designated TRICOM) induced the activation of T cells to a far greater extent than cells infected with vectors expressing any one or two costimulatory molecules. Despite this T-cell "hyperstimulation" using TRICOM vectors, no evidence of apoptosis above that seen using the B7-1 vector was observed. Results employing the TRICOM vectors were most dramatic under conditions of either low levels of first signal or low stimulator cell to T-cell ratios. Experiments employing a four-gene construct also showed that TRICOM recombinants could enhance antigen-specific T-cell responses in vivo. These studies thus demonstrate the ability of vectors to introduce three costimulatory molecules into cells, thereby activating both CD4+ and CD8+ T-cell populations to levels greater than those achieved with the use of only one or two costimulatory molecules. This new threshold of T-cell activation has broad implications in vaccine design and development. Dendritic cells infected with TRICOM vectors were found to greatly enhance naïve T-cell activation, and peptide-specific T-cell stimulation. In vivo, peptide-pulsed DCs infected with TRICOM vectors induced cytotoxic T lymphocyte activity markedly and significantly greater than peptide-pulsed DCs.


Assuntos
Transferência Adotiva , Células Dendríticas/transplante , Terapia Genética/métodos , Ativação Linfocitária/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Apresentação de Antígeno/imunologia , Apoptose/efeitos dos fármacos , Antígeno B7-1/administração & dosagem , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD58/administração & dosagem , Antígenos CD58/genética , Antígenos CD58/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Sinergismo Farmacológico , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Molécula 1 de Adesão Intercelular/administração & dosagem , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Poxviridae/genética
7.
Vaccine ; 20(5-6): 744-55, 2001 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-11738738

RESUMO

Since the rhesus is often used as a "gatekeeper" model for the evaluation of malaria and simian immunodeficiency virus (SIV)/HIV vaccines, the identification of strategies to enhance the activation of rhesus T cells would potentially aid in the generation of more potent vaccines directed against these infectious agents. Several molecules normally found on the surface of professional human APCs are capable of providing the second signals critical for T cell activation: B7-1 (CD80), ICAM-1 (CD54), and LFA-3 (CD58). With the exception of B7, T cell costimulatory molecules in the rhesus have not been identified. We have recently designed and characterized both recombinant vaccinia and recombinant avipox vectors containing the transgenes for a triad of human T cell costimulatory molecules (B7-1, ICAM-1, LFA-3; designated TRICOM). Here, we demonstrate the enhanced activation of rhesus T cells stimulated with rhesus APCs infected with TRICOM vectors in the presence of signal 1. Infection with TRICOM vectors led to significant improvement of APC capabilities in terms of reduction of the amount of signal 1 needed to activate naive T cells, and reduction in the amount of APCs required to activate T cells using a constant amount of signal 1. Antibody blocking studies demonstrated that each of the three costimulatory molecule transgenes contributed to the enhanced proliferation of T cells. TRICOM-enhanced T cell activation was shown to correspond to increases in type 1 cytokines and a reduced level of apoptosis. TRICOM-infected autologous B cells from rhesus immunized with either an SIV vaccine or a malaria vaccine stimulated significantly greater levels of IFN-gamma in response to specific peptide than stimulation with uninfected autologous B cells or B cells infected with wild-type vector. The ability to augment immune responses using poxvirus-based vaccines containing multiple costimulatory molecule transgenes can now be addressed in the rhesus macaque model.


Assuntos
Antígeno B7-1/genética , Antígenos CD58/genética , Molécula 1 de Adesão Intercelular/genética , Ativação Linfocitária , Linfócitos T/imunologia , Vacinas contra a AIDS/genética , Animais , Avipoxvirus/genética , Antígeno B7-1/administração & dosagem , Antígenos CD58/administração & dosagem , Linhagem Celular , Citocinas/biossíntese , Expressão Gênica , Vetores Genéticos , Humanos , Molécula 1 de Adesão Intercelular/administração & dosagem , Interferon gama/biossíntese , Ligantes , Macaca mulatta , Malária/imunologia , Vacinas contra a SAIDS/genética , Vaccinia virus/genética
8.
Cancer Biother Radiopharm ; 16(4): 305-15, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11603001

RESUMO

A phase I trial was designed to examine the feasibility of combining interferon and Taxol with intraperitoneal radioimmunotherapy (177Lu-CC49). Patients with recurrent or persistent ovarian cancer confined to the abdominal cavity after first line therapy, Karnofsky performance status > 60, adequate liver, renal and hematologic function, and tumor that reacted with CC49 antibody were enrolled. Human recombinant alpha interferon (IFN) was administered as 4 subcutaneous injections of 3 x 10(6) U on alternate days beginning 5 days before RIT to increase the expression of the tumor-associated antigen, TAG-72. The addition of IFN increased hematologic toxicity such that the maximum tolerated dose (MTD) of the combination was 40 mCi/m2 compared to 177Lu-CC49 alone (45 mCi/m2). Taxol, which has radiosensitizing effects as well as antitumor activity against ovarian cancer, was given intraperitoneally (i.p.) 48 hrs before RIT. It was initiated at 25 mg/m2 and escalated at 25 mg/m2 increments to 100 mg/m2. Subsequent groups of patients were treated with IFN + 100 mg/m2 Taxol + escalating doses of 177Lu-CC49. Three or more patients were treated in each dose group and 34 patients were treated with the 3-agent combination. Therapy was well tolerated with the expected reversible hematologic toxicity. The MTD for 177Lu-CC49 was 40 mCi/m2 when given with IFN + 100 mg/m2 Taxol. Interferon increased the effective whole body half-time of radioactivity and the whole body radiation dose. Taxol did not have a significant effect on pharmacokinetic or dosimetry parameters. Four of 17 patients with CT measurable disease had a partial response (PR) and 4 of 27 patients with non-measurable disease have progression-free intervals of 18+, 21+, 21+, and 37+ months. The combination of intraperitoneal Taxol chemotherapy (100 mg/m2) with RIT using 177Lu-CC49 and interferon was well tolerated, with bone marrow suppression as the dose-limiting toxicity.


Assuntos
Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ovarianas/terapia , Radioimunoterapia , Adenocarcinoma/diagnóstico por imagem , Adolescente , Adulto , Anticorpos Antineoplásicos/administração & dosagem , Feminino , Humanos , Injeções Intraperitoneais , Interferon Tipo I/administração & dosagem , Interferon Tipo I/farmacocinética , Lutécio/uso terapêutico , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias Ovarianas/diagnóstico por imagem , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Radioisótopos/uso terapêutico , Cintilografia , Proteínas Recombinantes , Resultado do Tratamento
9.
Cancer Res ; 61(20): 7568-76, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606396

RESUMO

Human dendritic cells (DCs) express MHC class I and II molecules and several T-cell costimulatory molecules that contribute to their efficiency as antigen-presenting cells (APCs). Whereas most human DC populations uniformly express some costimulatory molecules such as B7-2 (CD86), previous studies have shown a wide variation in the expression of B7-1 (CD80) among different human DC preparations. In the studies reported here, we demonstrate that replication-defective avipox vectors expressing B7-1 can be used to rapidly and efficiently infect human DCs and can enhance the efficacy of human DCs to activate specific human T-cell populations. This has been demonstrated both in systems using peptide as a source of signal 1 and in systems using recombinant avipox vector to deliver signal 1. The antigen used in these studies was the tumor-associated human carcinoembryonic antigen (CEA). An immunodominant 9-mer CTL epitope for CEA (designated CAP-1) has been previously characterized (K. Y. Tsang et al., J. Natl. Cancer Inst. (Bethesda), 87: 982-990, 1995). The source of signal 1 used in these studies was (a) the CAP-1 peptide; (b) recombinant avipox-CEA; or (c) the dual transgene recombinant avipox-CEA/B7-1. These studies demonstrate that CEA-specific T cells are more efficiently activated using as APCs peptide-pulsed DCs infected with avipox-B7-1, as compared with peptide-pulsed DCs infected with wild-type vector, or with uninfected peptide-pulsed DCs. Greater activation of CEA-specific T cells was also obtained using as APCs DCs that were infected with avipox-CEA/B7-1 as compared with the use of DCs infected with avipox-CEA. A CEA tetramer was also used to isolate high- and low-tetramer-binding CEA-specific T-cell populations. Although both high- and low-tetramer-binding T cells had the ability to lyse CEA peptide-pulsed targets, only the high-tetramer-binding T cells had the ability to lyse colon carcinoma cells expressing CEA, which suggests the existence of tetramer-binding populations with different T-cell receptor (TCR) affinities. The demonstrated safety of recombinant avipox vectors in humans and the previously demonstrated ability to administer them multiple times without host immune response limitations indicate that these vectors expressing B7-1 have a potential use in enhancing the efficacy of human DC immunotherapy protocols using either peptide or recombinant vector to deliver signal 1.


Assuntos
Avipoxvirus/genética , Antígeno B7-1/genética , Células Dendríticas/imunologia , Células Dendríticas/virologia , Epitopos de Linfócito T/imunologia , Linfócitos T Citotóxicos/imunologia , Apresentação de Antígeno/imunologia , Avipoxvirus/fisiologia , Antígeno B7-1/biossíntese , Antígeno B7-1/imunologia , Antígeno Carcinoembrionário/biossíntese , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Carcinoma/imunologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Expressão Gênica , Vetores Genéticos/genética , Humanos , Ativação Linfocitária/imunologia , Neoplasias Pélvicas/imunologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transgenes , Células Tumorais Cultivadas , Replicação Viral
10.
Cancer Res ; 61(14): 5517-22, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11454701

RESUMO

Surface-bound IgE play a central role in antiparasite immunity; to exploit IgE-driven immune mechanisms in tumor prevention and control, monoclonal IgEs of irrelevant specificity were loaded through biotin-avidin bridging onto tumor cells, either by systemic administration to tumor-bearing mice or pre-loading of tumor cells before inoculation. Here we show that systemic administration of biotinylated IgEs to mice bearing tumors pre-targeted with biotinylated antibodies and avidin significantly decreased tumor growth rate. In addition, as compared with IgG-loaded control cells, inoculation of suboptimal doses of IgE-loaded tumor cells suppressed tumor formation in a fraction of animals and induced protective host immunity by eliciting tumor-specific T-cell responses. Similarly, tumor vaccination experiments showed that irradiated tumor cells (IgE loaded by biotin-avidin bridging) conferred protective immunity at doses 100-fold lower than the corresponding control cells without IgE. Finally, in vivo depletion of eosinophils or T cells abrogated IgE-driven tumor growth inhibition. These results demonstrate that IgEs targeted on tumor cells not only possess a curative potential but also confer long-term antitumor immunity and that IgE-driven antitumor activity is not restricted to the activation of innate immunity effector mechanisms but also results from eosinophil-dependent priming of a T-cell-mediated adaptive immune response. This suggests a potential role for IgEs in the design of new cell-based tumor vaccines.


Assuntos
Anticorpos Monoclonais/farmacologia , Imunoglobulina E/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Divisão Celular/efeitos dos fármacos , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Feminino , Interleucina-5/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/prevenção & controle , Análise de Sobrevida , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/efeitos da radiação
11.
Cancer Biother Radiopharm ; 16(2): 133-46, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11385960

RESUMO

Among the radionuclides considered for radioimmunotherapy, alpha-emitters such as the bismuth isotopes, 212Bi and 213Bi, are of particular interest. The macrocyclic ligand, DOTA, has been shown to form stable complexes with bismuth isotopes. The kinetics of the complexation of bismuth with the DOTA chelate, however, are slow and impractical for use with 212Bi and 213Bi that have half-lives of 60.6 and 45.6 min. The study described herein compares six DTPA derived bifunctional chelates with the goal of identifying an alternative to the DOTA ligand for radiolabeling with bismuth. Radioimmunoconjugates comprised of MAb B72.3, each of the six DTPA chelates, and radiolabeled with 206Bi, which facilitated the evaluation due to its readily detectable gamma-emission. In vitro studies showed that each of the radioimmunoconjugates retained immunoreactivity that was comparable to its 125I-labeled counterpart. The 206Bi- and 125I-labeled immunoconjugates were then co-injected i.p. into normal athymic mice. Injection of Afree@ 206Bi demonstrated that the kidneys were the critical organ to evaluate for retention of bismuth in the chelate complex. Major differences were identified among the six preparations. The CHX-A and -B immunoconjugates were found to have 1) the lowest %ID/gm in the kidney; 2) a level of 206Bi in the kidney that was comparable to that of 125I-B72.3; and 3) no significant uptake of 206Bi evident in other organs such as bone, lung and spleen. The results described herein suggest that either of the cyclohexyl derivatives of DTPA may be suitable candidates for the labeling of immunoconjugates with alpha-emitting bismuth isotopes for radioimmunotherapeutic applications.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Bismuto/farmacocinética , Ácido Pentético/farmacocinética , Radioisótopos/farmacocinética , Animais , Quelantes , Feminino , Imunoconjugados , Rim/metabolismo , Camundongos , Camundongos Nus , Radioimunoensaio , Radioimunoterapia , Baço/metabolismo , Distribuição Tecidual
12.
Cancer Res ; 61(11): 4497-505, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389081

RESUMO

Several different vaccine strategies have been evaluated and combined in an attempt to amplify T-cell responses toward induction of antitumor immunity. The model tumor antigen used was carcinoembryonic antigen (CEA). While initial T-cell activation studies were conducted in conventional mice, combined vaccine strategy studies and antitumor studies were conducted in transgenic mice in which CEA is expressed in normal gastrointestinal tissue and CEA protein is found in sera. The studies reported here demonstrate: (a) A recombinant avipox (fowlpox, rF) vector expressing the signal 1 (CEA) and the B7-1 costimulatory molecule transgenes (designated rF-CEA/B7-1) is more potent in inducing CEA-specific T-cell responses than rF-CEA; one administration of recombinant fowlpox vector expressing CEA and three different costimulatory molecule transgenes (B7-1, ICAM-1, LFA-3, designated rF-CEA/TRICOM) was more potent in inducing CEA-specific T-cell responses than four vaccinations with rF-CEA or two vaccinations with rF-CEA/B7-1. Moreover, up to four vaccinations with rF-CEA/TRICOM induced greater CEA-specific T-cell responses with each vaccination. (b) A diversified prime and boost strategy using a prime with a recombinant vaccinia vector expressing CEA and the triad of costimulatory molecules (designated rV-CEA/TRICOM) and a boost with rF-CEA/TRICOM was more potent in inducing CEA-specific T-cell responses than the repeated use of rF-CEA/TRICOM alone. (c) The addition of granulocyte macrophage colony-stimulating factor (GM-CSF) to the rF-CEA or rF-CEA/TRICOM vaccinations via the simultaneous administration of a rF-GM-CSF vector enhanced CEA-specific T-cell responses. These strategies (TRICOM/diversified prime and boost/GM-CSF) were combined to treat CEA-expressing carcinoma liver metastases in CEA-transgenic mice; vaccination was initiated 14 days posttumor transplant. Antitumor effects in terms of survival and CD8(+) and CD4(+) responses specific for CEA were also observed in this CEA-transgenic mouse model. These studies demonstrate that the use of cytokines and diversified prime and boost regimens can be combined with the use of recombinant vectors expressing signal 1 and multiple costimulatory molecules to further amplify T-cell responses toward more effective vaccine strategies.


Assuntos
Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/imunologia , Neoplasias Hepáticas Experimentais/imunologia , Linfócitos T/imunologia , Animais , Vacinas Anticâncer/administração & dosagem , Antígeno Carcinoembrionário/genética , Epitopos de Linfócito T/imunologia , Feminino , Humanos , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
Crit Rev Oncol Hematol ; 39(1-2): 43-57, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11418301

RESUMO

Activation of T cells requires at least two signals: signal 1, via the T-cell receptor, and signal 2, in which a costimulatory molecule on the antigen presenting cell (APC) interacts with a ligand on the T cell. Dendritic cells (DCs) are the most potent APCs in part due to their expression of costimulatory molecules. DCs, however, constitute only a minor percentage of APCs in the body, and the in vitro preparation of DCs is both costly and time consuming. The studies reported here demonstrate that one can utilize other APCs, such as bone marrow progenitor cells (BMPCs) and make them markedly more effective as APCs; this was accomplished by their infection with recombinant poxviruses (either the replication-defective avipox or vaccinia), which contain transgenes for a triad of costimulatory molecules (B7-1, ICAM-1 and LFA-3, designated TRICOM). APCs infected with TRICOM vectors are shown to significantly enhance the activation of both naive and effector CD4(+) and CD8(+) T-cell populations. The use of TRICOM vectors in vaccine strategies is discussed.


Assuntos
Células Dendríticas/imunologia , Células-Tronco/efeitos dos fármacos , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Antígeno B7-1/genética , Antígeno B7-1/farmacologia , Células da Medula Óssea , Antígenos CD58/genética , Antígenos CD58/farmacologia , Células Dendríticas/efeitos dos fármacos , Feminino , Expressão Gênica , Vetores Genéticos/farmacologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Poxviridae/genética , Células-Tronco/imunologia , Células-Tronco/metabolismo
14.
Clin Cancer Res ; 7(5): 1181-91, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11350882

RESUMO

Granulocyte macrophage colony-stimulating factor (GM-CSF) has been shown to be an effective vaccine adjuvant because it enhances antigen processing and presentation by dendritic cells. ALVAC-CEA B7.1 is a canarypox virus encoding the gene for the tumor-associated antigen carcinoembryonic antigen (CEA) and for a T-cell costimulatory molecule, B7.1. After an initial dose escalation phase, this study evaluated vaccination with 4.5 x 10(8) plaque-forming units ALVAC-CEA B7.1 alone (n = 30) or with GM-CSF (n = 30) in patients with advanced CEA-expressing tumors to determine whether the addition of the adjuvant GM-CSF enhances induction of CEA-specific T-cells. Patients were vaccinated with vaccine intradermally every other week for 8 weeks. GM-CSF was given s.c. for 5 days beginning 2 days before vaccination. Patients with stable or responding disease after four immunizations received monthly boost injections alone or with GM-CSF. Biopsies of vaccine sites were obtained 48 h after vaccination to evaluate leukocytic infiltration and CEA expression. Induction of peripheral blood CEA-specific T-cell precursors was assessed in HLA-A2 positive patients by an ELISPOT assay looking for the production of IFN-gamma. Therapy was well tolerated. All of the patients had evidence of leukocytic infiltration and CEA expression in vaccine biopsy sites. In the patients receiving GM-CSF, leukocytic infiltrates were greater in cell number but were less likely to have a predominant lymphocytic infiltrate compared with patients receiving vaccine in the absence of the cytokine adjuvant. After four vaccinations, CEA-specific T-cell precursors were statistically increased in HLA-A2 positive patients who received vaccine alone. However, the GM-CSF plus vaccine cohort of HLA-A2 positive did not demonstrate a statistically significant increase in their CEA-specific T-cell precursor frequencies compared with baseline results. The number of prior chemotherapy regimens was negatively correlated with the generation of a T-cell response, whereas there was a positive correlation between the number of months from the last chemotherapy regimen and the T-cell response. ALVAC-CEA B7.1 is safe in patients with advanced, recurrent adenocarcinomas that express CEA, is associated with the induction of a CEA-specific T-cell response in patients treated with vaccine alone but not with vaccine and GM-CSF, and can lead to disease stabilization for up to 13 months.


Assuntos
Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias/tratamento farmacológico , Vacinas Sintéticas/uso terapêutico , Adulto , Idoso , Biópsia , Vacinas Anticâncer/efeitos adversos , Quimioterapia Adjuvante , Estudos de Coortes , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Humanos , Imunidade/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Resultado do Tratamento , Vacinas Sintéticas/efeitos adversos
15.
Vaccine ; 19(25-26): 3552-67, 2001 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-11348723

RESUMO

Recombinant orthopox vectors (both replication-defective fowlpox [rF], and replication competent vaccinia [rV] have been developed that simultaneously express three T-cell costimulatory molecule transgenes. The constituents of this triad of costimulatory molecules (designated TRICOM) are B7-1, ICAM-1, and LFA-3. We have previously shown that infection of murine dendritic cells (DCs) with TRICOM vectors increases their level of expression of the triad of costimulatory molecules and enhances the efficacy of DCs to activate T cells. While DCs are arguably the most potent antigen presenting cell (APC), limitations clearly exist in their use due to the level of effort and cost for their generation. The studies reported here demonstrate that a generic APC population, murine splenocytes, can be made markedly more efficient as APCs by infection with either rF-TRICOM or rV-TRICOM vectors. Infection of splenocytes with either TRICOM vector led to significant improvement of APC capabilities in terms of: (a) enhancement of mixed lymphocyte reactions; (b) a reduction in the amount of signal 1 to activate naive T cells; and (c) a reduction in the amount of APCs required to activate T cells using a constant amount of signal 1. TRICOM-enhanced T-cell activation was shown to correspond to increases in type-1 cytokines and a reduced level of apoptosis, compared with T cells activated with uninfected or control vector-infected splenocytes. In vitro and in vivo experiments compared DCs with TRICOM-infected splenocytes. Infection of splenocytes with TRICOM vectors markedly enhanced their ability to activate T cells to levels approaching that of DCs. These studies thus demonstrate for the first time that an abundant and accessible population of APCs obtainable without lengthy culture or the use of costly exogenous cytokines (in contrast to that of DCs) can be made more potent as APCs with the use of vectors that express a triad of costimulatory molecules.


Assuntos
Células Apresentadoras de Antígenos/imunologia , ISCOMs/administração & dosagem , ISCOMs/genética , Animais , Apoptose , Antígeno B7-1/administração & dosagem , Antígeno B7-1/genética , Linfócitos T CD4-Positivos/imunologia , Antígenos CD58/administração & dosagem , Antígenos CD58/genética , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Feminino , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos , Memória Imunológica , Molécula 1 de Adesão Intercelular/administração & dosagem , Molécula 1 de Adesão Intercelular/genética , Interferon gama/biossíntese , Interleucina-2/biossíntese , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Baço/citologia , Baço/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Vaccinia virus/genética
16.
Cancer Res ; 61(6): 2523-32, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11289125

RESUMO

The use of reverse transcription-PCR (RT-PCR) to analyze cells in the blood of cancer patients for the detection of mRNA expressed in tumor cells has implications for both the prognosis and the monitoring of cancer patients for the efficacy of established or experimental therapies. Carcinoembryonic antigen (CEA) is expressed on approximately 95% of colorectal, gastric, and pancreatic tumors, and on the majority of breast, non-small cell lung, and head and neck carcinomas. CEA shed in serum is useful as a marker in only approximately 50% of colorectal cancer patients and rarely is shed by some other carcinoma types. RT-PCR has been used previously to detect CEA mRNA in cells in the blood and lymph nodes of cancer patients. Under the assay conditions validated in the studies reported here, 34 of 51 (67%) patients with different stages of colorectal cancer had blood cells that were positive by RT-PCR for CEA mRNA, whereas none of 18 patients with colonic polyps were positive; 2 of 60 apparently healthy individuals (who were age and sex matched with the carcinoma patients and were part of a colon cancer screening program as controls) were marginally positive. The results of CEA PCR in the blood of the carcinoma patients and the other groups showed strong statistical correlation with the disease (P2 < 0.0001). Analyses were carried out to detect both serum CEA protein levels and CEA mRNA in blood cells of colorectal carcinoma patients by RT-PCR. For all stages of disease, 18 of 51 patients (35%) were positive for serum CEA, whereas 35 of 51 (69%) were positive by RT-PCR. More importantly, only 5 of 23 (20%) of stage B and C colorectal cancer patients were positive for serum CEA, whereas 16 of 23 (70%) were positive by RT-PCR. The use of two other serum markers (CA19.9 and CA72-4) for colorectal cancer in combination with serum CEA scored two additional patients as positive; both were positive by RT-PCR for CEA mRNA. Pilot long-term longitudinal studies conducted before and after surgery identified some patients with CEA mRNA in blood cells that were negative for all serum markers, who eventually developed clinical metastatic disease. The studies reported here are the first to correlate RT-PCR results for CEA mRNA in blood cells with one or more serum markers for patients with different stages of colorectal cancer, and are the first long-term longitudinal studies to use RT-PCR to detect CEA mRNA in blood cells of cancer patients. Larger cohorts will be required in future studies to define the impact, if any, of this technology on prognosis and/or disease monitoring.


Assuntos
Adenocarcinoma/sangue , Biomarcadores Tumorais/sangue , Antígeno Carcinoembrionário/genética , Neoplasias Colorretais/sangue , Células Neoplásicas Circulantes/imunologia , RNA Mensageiro/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos Glicosídicos Associados a Tumores/sangue , Antígenos Glicosídicos Associados a Tumores/genética , Biomarcadores Tumorais/genética , Antígeno CA-19-9/sangue , Antígeno CA-19-9/genética , Antígeno Carcinoembrionário/biossíntese , Antígeno Carcinoembrionário/sangue , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Feminino , Humanos , Imuno-Histoquímica , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Células Neoplásicas Circulantes/metabolismo , RNA Mensageiro/genética , Sensibilidade e Especificidade
17.
Cancer Res ; 61(9): 3725-34, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11325845

RESUMO

T-cell activation usually requires at least two signals. The first signal is antigen-specific, and the second signal(s) involves the interaction of a T-cell costimulatory molecule(s) on the antigen-presenting cell (APC) with its ligand on the T cell. Dendritic cells (DCs) are the most potent APCs, attributable, in part, to their expression of several T-cell costimulatory molecules. Human DCs generated in vitro, however, will vary in methods of generation and maturation and in terms of expression of different phenotypic markers-including costimulatory molecules-among different donors. We report here that a recombinant avipox (fowlpox, rF) vector has been constructed that can efficiently express the transgenes for three human T-cell costimulatory molecules (B7-1, ICAM-1, and LFA-3) as a result of individual early avipox promoters driving the expression of each transgene. This triad of costimulatory molecules (designated TRICOM) was selected because each has an individual ligand on T cells and each has been shown previously to prime a unique signaling pathway in T cells. We report here that rF-TRICOM can efficiently infect human DCs of different states of maturity and hyperexpress each of the three costimulatory molecules on the DC surface without affecting other DC phenotypic markers. Infection of influenza or human papilloma virus 9-mer peptide-pulsed DCs from different individuals, or at different stages of maturity with rF-TRICOM, resulted in enhanced activation of T cells from peripheral blood mononuclear cells of autologous donors after 24 h of incubation with DCS: This enhanced activation was analyzed by both titrating the peptide and differing the DC:effector cell ratios. No effect was observed using the control wild-type avipox vector. No increase in apoptosis was observed in T cells hyperstimulated with the TRICOM vector, and no decrease in interleukin-12 production was seen in lipopolysaccharide-stimulated DCs infected with rF-TRICOM. Antibody-blocking experiments demonstrated that enhanced T-cell activation by TRICOM was attributed to each of the three costimulatory molecules. Peptide-pulsed, rF-TRICOM-infected DCs were also shown to be more effective than peptide-pulsed DCs in activating T cells to 9-mer peptides derived from two relatively weak "self" immunogens, i.e., human prostate-specific antigen and human carcinoembryonic antigen. These studies thus demonstrate for the first time that a vector that can simultaneously hyperexpress three costimulatory molecules can be used to efficiently infect human DCs, leading to enhanced peptide-specific T-cell activation. The use of this approach for in vitro studies and clinical applications in immunotherapy is discussed.


Assuntos
Antígeno B7-1/imunologia , Antígenos CD58/imunologia , Células Dendríticas/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígenos CD58/biossíntese , Antígenos CD58/genética , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos/genética , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Interleucina-12/biossíntese , Células Tumorais Cultivadas
18.
J Clin Oncol ; 19(6): 1848-54, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11251017

RESUMO

There are major differences between therapeutic tumor vaccines and chemotherapeutic agents that have important implications for the design of early clinical trials. Many vaccines are inherently safe and do not require phase I dose finding trials. Patients with advanced cancers and compromised immune systems are not good candidates for assessing either the toxicity or efficacy of therapeutic cancer vaccines. The rapid pace of development of new vaccine candidates and the variety of possible adjuvants and modifications in method of administration makes it important to use efficient designs for clinical screening and evaluation of vaccine regimens. We review the potential advantages of a wide range of clinical trial designs for the development of tumor vaccines. We address the role of immunological endpoints in early clinical trials of tumor vaccines, investigate the design implications of attempting to use disease stabilization as an end point and discuss the difficulties of reliably utilizing historical control data. Several conclusions for expediting the clinical development of effective cancer vaccines are proposed.


Assuntos
Antineoplásicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Determinação de Ponto Final , Neoplasias/virologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Adjuvantes Imunológicos/uso terapêutico , Estudos de Coortes , Humanos , Hospedeiro Imunocomprometido , Neoplasias/tratamento farmacológico , Projetos de Pesquisa , Resultado do Tratamento
19.
Crit Rev Oncol Hematol ; 38(1): 3-16, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11255077

RESUMO

Monoclonal antibody (MAb) CC49 reacts with a pancarcinoma antigen, tumor associated glycoprotein (TAG)-72. To circumvent human anti-murine antibody (HAMA) responses in patients, we earlier developed a humanized CC49 (HuCC49) by grafting the complementarity-determining regions (CDRs) of MAb CC49 onto variable light (VL) and variable heavy (VH) frameworks of the human MAbs LEN and 21/28'CL, respectively. With the aim of minimizing its immunogenicity further, we have now generated a variant HuCC49 MAb by grafting the specificity-determining residues (SDRs) of MAb CC49 onto the frameworks of the human MAbs. Based on the evaluation of its binding affinity for TAG-72 and its reactivity with anti-idiotypic antibodies present in sera from patients who have been treated with murine CC49, this variant retains its antigen-binding activity and shows minimal reactivity with anti-idiotypic antibodies in patients' sera. Development of this variant, which is a potentially useful clinical reagent for diagnosis and therapy of human carcinomas, demonstrates that for humanization of a xenogeneic antibody grafting of the potential SDRs should be sufficient to retain its antigen-binding properties.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Anticorpos Heterófilos/genética , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Regiões Determinantes de Complementaridade/análise , Regiões Determinantes de Complementaridade/genética , Desenho de Fármacos , Engenharia Genética , Variação Genética , Humanos
20.
Cancer Res ; 61(1): 206-14, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11196163

RESUMO

Recombinant avian poxviruses [fowlpox and canarypox (ALVAC)], restricted for replication in nonavian cell substrates and expressing granulocyte/macrophage-colony stimulating factor (avipox-GM-CSF), were evaluated for their ability to enrich an immunization site with antigen-presenting cells (APCs) and, in turn, function as biological vaccine adjuvants. Avipox-GM-CSF administered as a single s.c. injection significantly enhanced the percentage and absolute number of APCs in the regional lymph nodes that drain the injection site. Both the magnitude and duration of the cellular and phenotypic increases within the lymph nodes induced by the avipox-GM-CSF viruses were significantly (P < 0.05) greater than those measured in mice treated with four daily injections of recombinant GM-CSF protein. Temporal studies revealed that the APC enrichment of regional lymph nodes was sustained for 21-28 days after injection of the recombinant avipox virus expressing GM-CSF and, moreover, three injections of the recombinant virus could be given without any appreciable loss of in vivo bioactivity. Mice expressing human carcinoembryonic antigen (CEA) as a transgene (CEA.Tg) developed CEA-specific humoral and cell-mediated immunity after being immunized with avipox-CEA. The coadministration of recombinant avipox viruses expressing CEA and GM-CSF significantly enhanced CEA-specific host immunity with an accompanying immunotherapeutic response in tumor-bearing CEA.Tg mice. The optimal use of avipox-GM-CSF, in terms of dose and dose schedule, especially when used with different immunogens, remains to be determined. Nonetheless, the present findings demonstrate: (a) the effective delivery of GM-CSF to an immunization site using a recombinant avian poxvirus; (b) the compatibility of delivering an antigen and GM-CSF in replication-defective viruses to enhance antigen-specific immunity; and (c) the combined use of recombinant avipox viruses expressing CEA and GM-CSF to generate antitumor immunity directed at a self tumor antigen.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Linfonodos/imunologia , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antineoplásicos/biossíntese , Anticorpos Antineoplásicos/sangue , Avipoxvirus/genética , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Embrião de Galinha , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Linfonodos/citologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Proteínas Recombinantes , Linfócitos T/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...