Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Clin Cancer Res ; 24(16): 3845-3856, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30018119

ABSTRACT

Purpose: Dendritic cells (DC) initiate adaptive immune responses through the uptake and presentation of antigenic material. In preclinical studies, intratumorally injected activated DCs (aDCs; DCVax-Direct) were superior to immature DCs in rejecting tumors from mice.Experimental Design: This single-arm, open-label phase I clinical trial evaluated the safety and efficacy of aDCs, administered intratumorally, in patients with solid tumors. Three dose levels (2 million, 6 million, and 15 million aDCs per injection) were tested using a standard 3 + 3 dose-escalation trial design. Feasibility, immunogenicity, changes to the tumor microenvironment after direct injection, and survival were evaluated. We also investigated cytokine production of aDCs prior to injection.Results: In total, 39 of the 40 enrolled patients were evaluable. The injections of aDCs were well tolerated with no dose-limiting toxicities. Increased lymphocyte infiltration was observed in 54% of assessed patients. Stable disease (SD; best response) at week 8 was associated with increased overall survival. Increased secretion of interleukin (IL)-8 and IL12p40 by aDCs was significantly associated with survival (P = 0.023 and 0.024, respectively). Increased TNFα levels correlated positively with SD at week 8 (P < 0.01).Conclusions: Intratumoral aDC injections were feasible and safe. Increased production of specific cytokines was correlated with SD and prolonged survival, demonstrating a link between the functional profile of aDCs prior to injection and patient outcomes. Clin Cancer Res; 24(16); 3845-56. ©2018 AACR.


Subject(s)
Cell- and Tissue-Based Therapy , Dendritic Cells/transplantation , Immunotherapy , Neoplasms/therapy , Adult , Aged , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/immunology , Female , Humans , Injections, Intralesional , Interleukin-12/genetics , Interleukin-8/genetics , Male , Middle Aged , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
J Transl Med ; 1(1): 7, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-14580262

ABSTRACT

Dendritic cells (DC) are increasingly prepared in vitro for use in immunotherapy trials. Mature DC express high levels of surface molecules needed for T cell activation and are superior at antigen-presentation than immature DC. Bacillus Calmette-Guerin (BCG) is one of several products known to induce DC maturation, and interferon (IFN)-gamma has been shown to enhance the activity of DC stimulated with certain maturation factors. In this study, we investigated the use of IFN-gamma in combination with the powerful maturation agent, BCG. The treatment of immature DC with IFN-gamma plus BCG led to the upregulation of CD54, CD80, and CD86 in comparison with BCG treatment alone. In MLR or recall immune responses, the addition of IFN-gamma at the time of BCG-treatment did not increase the number of antigen-specific T cells but enhanced the development of IFN-gamma-producing Th1 cells. In primary immune responses, on the other hand, BCG and IFN-gamma co-treated DC stimulated higher proportions of specific T cells as well as IFN-gamma secretion by these T cells. Thus the use of IFN-gamma during BCG-induced DC maturation differentially affects the nature of recall versus naïve antigen-specific T-cell responses. IFN-gamma co-treatment with BCG was found to induce IL-12 and, in some instances, inhibit IL-10 secretion by DC. These findings greatly enhance the potential of BCG-matured dendritic cells for use in cancer immunotherapy.

4.
Cancer Immunol Immunother ; 52(7): 413-22, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12835918

ABSTRACT

Active immunotherapy of cancer requires the availability of a source of tumor antigens. To date, no such antigen associated with lung cancer has been identified. We have therefore investigated the ability of dendritic cells (DC) to capture whole irradiated human lung tumor cells and to present a defined surrogate antigen derived from the ingested tumor cells. We also describe an in vitro system using a modified human adenocarcinoma cell line (A549-M1) that expresses the well-characterized, immunogenic influenza M1 matrix protein as a surrogate tumor antigen. Peripheral blood monocyte-derived DC, when co-cultured with sub-lethally irradiated A549 cells or primary lung tumor cells derived from surgical resection of non-small cell carcinoma (NSCLC), efficiently ingested the tumor cells as determined by flow cytometry analysis and confocal microscopic examination. More importantly, DC loaded with irradiated A549-M1 cells efficiently processed and presented tumor cell-derived M1 antigen to T cells and elicited antigen-specific immune responses that included IFNgamma release from an M1-specific T-cell line, expansion of M1 peptide-specific Vbeta17+ and CD8+ peripheral T cells and generation of M1-specific cytotoxic T lymphocytes (CTL). We also compared DC loaded with irradiated tumor cells to those loaded with tumor cell lysate or killed tumor cells and found that irradiated lung tumor cells as a source of tumor antigen for DC loading is superior to tumor cell lysate or killed tumor cells in efficient induction of antigen-specific T-cell responses. Our results demonstrate the feasibility of using lung tumor cell-loaded DC to induce immune responses against lung cancer-associated antigens and support ongoing efforts to develop a DC-based lung cancer vaccine.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/physiology , Lung Neoplasms/immunology , T-Lymphocytes/immunology , Adenocarcinoma/immunology , Adenoviridae/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/immunology , Coculture Techniques , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Humans , Influenza A virus/immunology , Lymphocyte Activation , Microscopy, Confocal , Tumor Cells, Cultured/radiation effects , Viral Matrix Proteins/immunology
5.
Virology ; 308(1): 178-90, 2003 Mar 30.
Article in English | MEDLINE | ID: mdl-12706101

ABSTRACT

Although most HIV-1 infections worldwide result from heterosexual transmission, most vaccine candidates have focused on induction of systemic immunity and protection. We hypothesized that combining systemic priming with mucosal boosting would induce mucosal immunity that would protect from intravaginal challenge. Macaques were primed systemically with recombinant vaccinia viruses and boosted mucosally using inactivated SHIV(89.6) plus adjuvant. Other animals received protein boosts with adjuvant alone. Priming and boosting induced antiviral IgG and IgA antibodies. Such antibodies were induced to a lesser degree in animals receiving boosts alone. Anti-SHIV T cell responses were induced only in the prime-boost animals. Immunized animals and controls were challenged intravaginally with SHIV(89.6) and significant reductions in proviral and viral RNA loads were observed in the prime-boost animals. The boost-only animals did not have significant viral load reductions. These data suggest that cellular immunity was required for protection from intravaginal challenge. This immunization regimen provides a promising lead for vaccine development.


Subject(s)
AIDS Vaccines/administration & dosage , HIV Infections/immunology , Lentiviruses, Primate/immunology , Virus Replication/immunology , Administration, Cutaneous , Administration, Intranasal , Animals , Antibodies, Viral/blood , DNA, Viral/analysis , Disease Models, Animal , Female , HIV Infections/blood , HIV Infections/prevention & control , Immunization/methods , Immunoglobulin A/blood , Immunoglobulin G/blood , Macaca nemestrina , Proviruses/genetics , RNA, Viral/blood , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Viral Load
6.
J Immunother ; 25(4): 289-303, 2002.
Article in English | MEDLINE | ID: mdl-12142552

ABSTRACT

The immunotherapy of cancer is predicated on the belief that it is possible to generate a clinically meaningful antitumor response that provides patient benefit, such as improvement in the time to progression or survival. Indeed, immunotherapeutics with dendritic cells (DC) as antigen-presenting delivery vehicles for cell-based vaccines have already improved patient outcome against a wide range of tumor types (1-9). This approach stimulates the patient's own antitumor immunity through the induction or enhancement of T-cell immunity. It is generally believed that the activity of cytotoxic T lymphocytes (CTL), the cells directly responsible for killing the tumor cells in vivo, are directed by DC. Therefore, the goal of many current designs for DC-based vaccines is to induce strong tumor-specific CTL responses in patients with cancer. In practice, most studies for DC-based cancer vaccine development have focused on the development of methods that can effectively deliver exogenous tumor antigens to DC for cross-priming of CD8+ T cells through the endogenous MHC class I processing and presentation pathway (10). To date, many methods have been developed or evaluated for the delivery of defined and undefined tumor antigens to DC. This review provides a brief summary on these methods, the techniques used in these methods, as well as the advantages and disadvantages of each method.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Animals , Bacterial Toxins/immunology , DNA, Neoplasm/immunology , Humans , Immunity, Active/immunology , Liposomes , Peptides/immunology , Proteins/immunology , RNA, Neoplasm/immunology , Receptors, IgG/immunology , Recombinant Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...