Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Gynecol Oncol ; 170: 221-228, 2023 03.
Article in English | MEDLINE | ID: mdl-36709663

ABSTRACT

BACKGROUND: IGF signaling has been implicated in the pathogenesis and progression of ovarian carcinoma (OC). Single agent activity and safety of ganitumab (AMG 479), a fully human monoclonal antibody against IGF1R that blocks binding of IGF1 and IGF2, were evaluated in patients with platinum-sensitive recurrent OC. METHODS: Patients with CA125 progression (GCIG criteria) or measurable disease per RECIST following primary platinum-based therapy received 18 mg/kg of ganitumab q3w. The primary endpoint was objective response rate (ORR) assessed per RECIST 1.1 by an independent radiology review committee (IRC) and/or GCIG CA125 criteria. Secondary endpoints included clinical benefit rate (CBR), progression free survival (PFS) and overall survival (OS). RESULTS: 61 pts. were accrued. Objective responses were seen in 5/61 patients (ORR 8.2%, 95% CI, 3.1-18.8) with 1 partial response (PR) by RECIST and 2 complete responses (CR) as well as 2 PR by CA125 criteria. CBR was 80.3% (95% CI, 67.8-89.0%). The median PFS according to RECIST by IRC was 2.1 months (95% CI, 2.0-3.1). The median PFS per RECIST IRC and/or CA125 was 2.0 months (95% CI, 1.8-2.2). The median OS was 21 months (95% CI, 19.5-NA). The most common overall adverse events were fatigue (36.1%) and hypertension (34.4%). Grade 1/2 hyperglycemia occurred in 30.4% of patients. Hypertension (11.5%) and hypersensitivity (8.2%) were the most frequent grade 3 adverse events. CONCLUSIONS: IGF1R inhibition with ganitumab was well-tolerated, however, our results do not support further study of ganitumab as a single agent in unselected OC patients.


Subject(s)
Antibodies, Monoclonal, Humanized , Ovarian Neoplasms , Humans , Female , Antibodies, Monoclonal/adverse effects , Ovarian Neoplasms/drug therapy
2.
Gynecol Oncol ; 163(3): 465-472, 2021 12.
Article in English | MEDLINE | ID: mdl-34642026

ABSTRACT

PURPOSE: Insulin-like growth factor (IGF) signaling is implicated in pathogenesis and chemotherapy resistance of epithelial ovarian cancer (EOC). We explored efficacy and safety of adding ganitumab, a monoclonal antibody targeting IGF-1R, to carboplatin/paclitaxel (CP) chemotherapy in patients with primary EOC. DESIGN: Patients were randomly assigned to receive CP/ganitumab (18 mg/kg q3w) or CP/placebo for 6 cycles followed by 6 cycles of single agent ganitumab/placebo maintenance therapy as front-line therapy. Primary endpoint was progression free survival. Secondary endpoints were time to progression and overall survival. Pretreatment samples were prospectively collected for retrospective biomarker analyses. RESULTS: 170 patients enrolled. 165 patients assessable for toxicity. Median PFS was 15.7 months with CP/ganitumab and 16.7 months with CP/placebo (HR 1.23; 95% CI 0.82-1.83, P = 0.313). All grade neutropenia (84.1% vs 71.4%), thrombocytopenia (75.3% vs 57.1%) and hyperglycemia (15.9% vs 2.6%) were more common in the ganitumab group compared to the placebo group. Ganitumab/placebo related serious adverse events were reported in 26.1% of the patients with ganitumab and in 6.5% with placebo. Non-progression related fatal events were more common with ganitumab (5 versus 2 patients). The ganitumab group experienced more dose delays which resulted in lower relative dose intensity of chemotherapy in the experimental group. In an exploratory model IGFBP2 expression was predictive of ganitumab response (treatment interaction; PFS, P = 0.03; OS, P = 0.01). CONCLUSION: Addition of ganitumab to CP chemotherapy in primary EOC did not improve PFS. Our results do not support further study of ganitumab in unselected EOC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Ovarian Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/metabolism , Carboplatin/administration & dosage , Carboplatin/adverse effects , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Insulin-Like Growth Factor Binding Protein 2/metabolism , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Progression-Free Survival , Somatomedins/metabolism
3.
Br J Cancer ; 87(3): 268-76, 2002 Jul 29.
Article in English | MEDLINE | ID: mdl-12177793

ABSTRACT

In part A of this study, patients were randomised to cohorts receiving darbepoetin alfa at doses of 0.5 to 8.0 m.c.g x kg(-1) x wk(-1) or to a control group receiving epoetin alfa at an initial dose of 150 U x kg(-1) three times weekly. In part B, the cohorts were darbepoetin alfa 3.0 to 9.0 m.c.g x kg(-1) every 2 weeks or epoetin alfa, initial dose 40 000 U x wk(-1). Safety was assessed by adverse events, changes in blood pressure, and formation of antibodies to darbepoetin alfa. Efficacy was assessed by several haematologic endpoints, including change in haemoglobin from baseline. The adverse event profile of darbepoetin alfa was similar to that of epoetin alfa. No relationship between the rapidity of haemoglobin response and any adverse event was observed. No antibodies to darbepoetin alfa were detected. Higher doses of darbepoetin alfa increased the proportion of patients with a haemoglobin response and decreased the median time to response. The overall dose of darbepoetin alfa required to produce a mean increase in haemoglobin does not increase when the dosing interval is increased from 1 to 2 weeks. Therapy with darbepoetin alfa is safe and effective in producing a dose-related increase in haemoglobin levels in patients with cancer receiving chemotherapy.


Subject(s)
Anemia/drug therapy , Antineoplastic Agents/adverse effects , Erythropoietin/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Darbepoetin alfa , Dose-Response Relationship, Drug , Erythropoietin/administration & dosage , Erythropoietin/adverse effects , Erythropoietin/analogs & derivatives , Female , Hemoglobins/analysis , Humans , Male , Middle Aged , Neoplasms/blood , Neoplasms/complications , Recombinant Proteins
4.
Leukemia ; 16(8): 1484-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145689

ABSTRACT

The ability of interferon-alpha (IFN-alpha) to induce dendritic cell (DC) differentiation in chronic myeloid leukemia (CML) was evaluated. Peripheral blood mononuclear cells from CML patients cultured with IFN-alpha and granulocyte-macrophage colony-stimulating factor (GM-CSF) developed a dendritic morphology. Fluorescence in situ hybridization demonstrated that the DCs harbored the bcr/abl translocation. The DCs prepared with IFN-alpha/GM-CSF expressed significantly higher levels of class I and II HLA than those grown in interleukin-4 (IL-4) and GM-CSF. The DCs prepared from newly diagnosed CML patients using IFN-alpha/GM-CSF expressed immunoregulatory proteins at levels comparable to normal DCs. In contrast, DCs cultured from CML patients who did not achieve a cytogenetic response to IFN-alpha expressed significantly lower levels of class I HLA, CD40, CD54, CD80 and CD86 than normal DCs. The expression of CD86 by CML DCs was enhanced when they were cultured with IFN-alpha/IL-4/GM-CSF, or when IFN-alpha/GM-CSF-treated cells were induced to mature by CD40 ligand. The DCs from IFN-alpha failures were less stimulatory than normal DCs in the allogeneic mixed leukocyte reaction. CML patients who had a cytogenetic response to IFN-alpha initially had low numbers of bone marrow DCs that increased significantly with treatment, while nonresponders had more prevalent DCs at baseline that showed no consistent change with treatment. Therefore, IFN-alpha can induce DC differentiation from CML progenitor cells both in vitro and in vivo. The therapeutic activity of IFN-alpha in CML may be due to its ability to stimulate the generation of DCs that can present CML-specific antigens. Resistance to IFN-alpha may result when DC differentiation becomes impaired.


Subject(s)
Dendritic Cells/drug effects , Interferon-alpha/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/drug effects , Antigen Presentation , Antigens, CD/analysis , Antigens, CD/biosynthesis , Antigens, CD/genetics , Biomarkers, Tumor/genetics , Blood Cells/pathology , Bone Marrow Cells/pathology , CD40 Ligand/pharmacology , Cell Differentiation/drug effects , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA Antigens/analysis , HLA Antigens/biosynthesis , HLA Antigens/genetics , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Interferon alpha-2 , Lymphocyte Culture Test, Mixed , Neoplastic Stem Cells/pathology , Recombinant Proteins , Tumor Cells, Cultured/drug effects
5.
Cancer Res ; 61(24): 8782-6, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751399

ABSTRACT

alpha-Fetoprotein (AFP) is a potential target for immunotherapy in hepatocellular carcinoma; both the murine and human T-cell repertoires can recognize AFP-derived epitopes in the context of the MHC. Protective immunity can be generated with AFP-engineered dendritic cell-based vaccines. We now report a DNA-based immunization strategy using a prime-boost approach: coadministration of plasmid DNA encoding murine AFP and murine granulocyte-macrophage colony-stimulating factor followed by boosting with an AFP-expressing nonreplicating adenoviral vector. This immunization strategy can elicit a high frequency of Th1-type AFP-specific cells leading to tumor protective immunity in mice at levels comparable with AFP-engineered dendritic cells. This cell-free mode of immunization is better suited for large-scale vaccine efforts for patients with hepatocellular carcinoma.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoma, Hepatocellular/therapy , Immunodominant Epitopes/immunology , Liver Neoplasms/therapy , alpha-Fetoproteins/genetics , alpha-Fetoproteins/immunology , Adenoviridae/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Epitopes, T-Lymphocyte/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunodominant Epitopes/genetics , Immunotherapy, Active/methods , Jurkat Cells/immunology , Jurkat Cells/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , Plasmids/genetics , T-Lymphocytes/immunology , Transfection , Vaccines, DNA/genetics , Vaccines, DNA/immunology , alpha-Fetoproteins/biosynthesis
6.
Cancer Res ; 61(24): 8787-93, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751400

ABSTRACT

Genetic immunization of mice with dendritic cells (DCs) engineered to express a melanoma antigen generates antigen-specific, MHC-restricted, CD4-dependent protective immune responses. We wanted to determine the role of CD4 cells and CD40 ligation of MART-1 gene-modified DC in an animal model of immunotherapy for murine melanoma. CD4 knock-out (CD4KO) or antibody-depleted mice were immunized with DC adenovirally transduced with the MART-1 gene (AdVMART1/DC) with or without CD40 cross-linking. Tumor protection was absent in CD4-depleted mice, but protection was reestablished when the CD40 receptor was engaged using three different constructs. Transduction of DCs with vectors expressing the Th1 cytokines interleukin (IL)-2, IL-7, or IL-12 could not reproduce the CD40-mediated maturation signal in this model. CD8 T-cell depletion in CD4KO mice immunized with CD40-ligated DCs abrogated the protective response. Pooled analysis of CD40 cross-linking of AdVMART1/DC administered to wild-type C57BL/6 mice revealed an overall enhancement of antitumor immunity. However, this effect was inconsistent between replicate studies. In conclusion, maturation of AdVMART1-transduced DCs through the CD40 ligation pathway can promote a protective CD8 T-cell-mediated immunity that is independent of CD4 T-cell help.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Adenoviridae/genetics , Animals , Antigens, Neoplasm , CD40 Antigens/genetics , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Epitopes, T-Lymphocyte/immunology , Genetic Vectors/genetics , Interleukins/biosynthesis , Interleukins/genetics , Interleukins/immunology , Lymphoma/immunology , Lymphoma/therapy , MART-1 Antigen , Melanoma, Experimental/prevention & control , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Transduction, Genetic
7.
Urology ; 58(2): 283-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489728

ABSTRACT

OBJECTIVES: The results of epidemiologic and animal studies support the role of a low-fat diet supplemented with omega-3 fatty acids contained in fish oil in preventing the development and progression of prostate cancer. As a first step in studying the role of a low-fat, fish oil-supplemented (LF/FOS) diet in a clinical setting, we conducted a prospective study in men with untreated prostate cancer to evaluate whether a 3-month dietary intervention affects the ratio of omega-3 to omega-6 fatty acids in plasma and gluteal fat. In addition, we evaluated the feasibility of studying cyclooxygenase-2 (COX-2) expression in serial prostate needle biopsy specimens before and after the diet. METHODS: Nine men with untreated prostate cancer consumed an LF/FOS diet for 3 months. Plasma, gluteal adipose tissue, and prostate needle biopsy specimens were obtained from each patient before and after the intervention. The fatty acid compositions of the plasma and gluteal adipose tissue were determined by gas-liquid chromatography, and the COX-2 expression in the prostatic tissue specimens was determined by semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: Short-term intervention with an LF/FOS diet caused a significant increase in the omega-3/omega-6 fatty acid ratio in plasma (P = 0.002) and gluteal adipose tissue (P = 0.002) in men with prostate cancer. The COX-2 expression in prostatic tissue was quantitated by RT-PCR in 7 of 9 patients, and COX-2 expression decreased in 4 of these 7 patients. CONCLUSIONS: A short-term dietary intervention in men with prostate cancer leads to a significant increase in the omega-3/omega-6 fatty acid ratios in plasma and adipose tissue. The potential for this diet to prevent the development and progression of prostate cancer by way of altered COX-2 expression and prostaglandin production in prostatic tissue requires further study.


Subject(s)
Diet, Fat-Restricted , Fatty Acids, Omega-3/administration & dosage , Fish Oils/administration & dosage , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/prevention & control , Adenocarcinoma , Adipose Tissue/chemistry , Aged , Aged, 80 and over , Biopsy, Needle , Buttocks/pathology , Cyclooxygenase 2 , Dietary Supplements , Disease Progression , Fatty Acids, Omega-3/blood , Humans , Male , Membrane Proteins , Middle Aged , Neoplasm Staging , Prospective Studies , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/diet therapy , Prostatic Neoplasms/pathology
8.
J Immunol ; 166(8): 5300-8, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290817

ABSTRACT

alpha fetoprotein (AFP)-derived peptide epitopes can be recognized by human T cells in the context of MHC class I. We determined the identity of AFP-derived peptides, presented in the context of HLA-A*0201, that could be recognized by the human (h) T cell repertoire. We screened 74 peptides and identified 3 new AFP epitopes, hAFP(137-145), hAFP(158-166), and hAFP(325-334), in addition to the previously reported hAFP(542-550.) Each possesses two anchor residues and stabilized HLA-A*0201 on T2 cells in a concentration-dependent class I binding assay. The peptides were stable for 2-4 h in an off-kinetics assay. Each peptide induced peptide-specific T cells in vitro from several normal HLA-A*0201 donors. Importantly, these hAFP peptide-specific T cells also were capable of recognizing HLA-A*0201(+)/AFP(+) tumor cells in both cytotoxicity assays and IFN-gamma enzyme-linked immunospot assays. The immunogenicity of each peptide was tested in vivo with HLA-A*0201/K(b)-transgenic mice. After immunization with each peptide emulsified in CFA, draining lymph node cells produced IFN-gamma on recognition of cells stably transfected with hAFP. Furthermore, AFP peptide-specific T cells could be identified in the spleens of mice immunized with dendritic cells transduced with an AFP-expressing adenovirus (AdVhAFP). Three of four AFP peptides could be identified by mass spectrometric analysis of surface peptides from an HLA-A*0201 human hepatocellular carcinoma (HCC) cell line. Thus, compelling immunological and physiochemical evidence is presented that at least four hAFP-derived epitopes are naturally processed and presented in the context of class I, are immunogenic, and represent potential targets for hepatocellular carcinoma immunotherapy.


Subject(s)
HLA-A2 Antigen/immunology , Lymphocyte Activation , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , alpha-Fetoproteins/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Alleles , Animals , Antigen Presentation/genetics , Cell Line, Transformed , Cells, Cultured , Cytotoxicity Tests, Immunologic , Dendritic Cells/transplantation , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , H-2 Antigens/genetics , HLA-A2 Antigen/genetics , Humans , Jurkat Cells , K562 Cells , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , alpha-Fetoproteins/administration & dosage , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
9.
Cancer Res ; 60(22): 6457-64, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103813

ABSTRACT

The cytokine interleukin-12 (IL-12) has shown potent antitumor activity in several tumor models. Recently, natural killer (NK) T cells have been proposed to mediate the antitumor effects of IL-12. In this study, the antitumor response of IL-12 was investigated in a gene therapeutic model against s.c. growing mouse hepatocellular carcinomas using an adenoviral vector expressing murine IL-12 (AdVmIL-12). An adenoviral-based system was chosen because of the ability of adenoviruses to transduce dividing and nondividing cells and because of their high transduction efficiencies. Our goals were to examine the efficacy of AdVmIL-12 in a hepatocellular carcinoma model and to investigate the mechanism of the AdVmIL-12-mediated antitumor response with specific interest in the role of NK T cells. Our studies demonstrate that intratumoral AdVmIL-12-mediated regression of s.c. hepatocellular tumors is associated with rapid antitumor responses. AdVmIL-12 treatment was associated with an immune cellular infiltrate consisting of CD4 and CD8 T lymphocytes, macrophages, NK cells, and NK T cells. Antibody ablation of CD4 and CD8 T cells and use of NK cell-defective beige mice failed to abrogate the response to AdVmIL-12. Studies in T-cell- and B-cell-deficient severe combined immunodeficient and recombinase activating gene-2-deficient mice and T-cell-, B-cell-, and NK cell-defective severe combined immunodeficient/beige mice also failed to abrogate this response. AdVmIL-12 retained potent antitumor activity in mice with specific genetic defects in immune cellular cytotoxicity (perforin knockout mice) and costimulation (CD28 knockout mice). Use of mice with specific NK T cell deficiencies, Valpha14 T-cell receptor and CD1 knockout mice, also failed to abrogate the response to AdVmIL-12. Histological and immunohistochemical studies of AdVmIL-12-treated tumors showed extensive inhibition of neovascularization and a marked decrease in factor VIII-stained endothelial cells. Our studies indicate that the antitumor response of AdVmIL-12 is independent of direct cytotoxic cellular immunity (specifically, the function of NK T cells) and suggest that the initial mechanisms of AdVmIL-12-mediated tumor regression involve inhibition of angiogenesis.


Subject(s)
Antigens, CD1/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD28 Antigens/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunocompromised Host/immunology , Interleukin-12/genetics , Liver Neoplasms, Experimental/therapy , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neovascularization, Pathologic/prevention & control , Perforin , Pore Forming Cytotoxic Proteins
10.
Blood ; 96(7): 2385-90, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11001888

ABSTRACT

The safety and efficacy of administering ex vivo expanded peripheral blood progenitor cells (PBPC) to patients with breast cancer who undergo high-dose chemotherapy and PBPC transplantation was investigated. Unselected PBPC were cultured in gas-permeable bags containing 1-L serum-free media, granulocyte colony-stimulating factor, stem cell factor, and pegylated megakaryocyte growth and development factor for 9 days. Cell dose cohorts were assigned to have between 2 and 24 x 10(9) PBPC cultured at 1, 2, or 3 x 10(6) cells/mL. Twenty-four patients received high-dose chemotherapy followed by infusion of the cultured PBPC and at least 5 x 10(6) CD34(+) uncultured cryopreserved PBPC per kilogram. No toxicities resulted from infusions of the ex vivo expanded PBPC. The study patients had shorter times to neutrophil (P =.0001) and platelet (P =.01) recovery and fewer red cell transfusions (P =.02) than 48 historical controls who received the same conditioning regimen and posttransplantation care and at least 5 x 10(6) CD34(+) PBPC per kilogram. Improvements in all these endpoints were significantly correlated with the expanded cell dose. Nine of 24 (38%) patients recovered neutrophil counts above 500/microL by day 5 or 6 after transplantation, whereas none of the controls had neutrophil recovery before the eighth day. Seven (29%) patients had neutropenia for 3 or fewer days, and 9 (38%) patients did not experience neutropenic fevers or require broad-spectrum antibiotics. Therefore, ex vivo expanded PBPC are capable of ameliorating posttransplantation neutropenia, thrombocytopenia, and anemia in patients receiving high-dose chemotherapy.


Subject(s)
Anemia/prevention & control , Breast Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Neutropenia/prevention & control , Thrombocytopenia/prevention & control , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Cell Count , Cells, Cultured , Culture Media, Serum-Free , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Length of Stay , Platelet Transfusion , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology
13.
Cancer Res ; 60(8): 2218-24, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10786687

ABSTRACT

Genetic immunization with a single injection of dendritic cells (DCs) expressing a model melanoma antigen generates antigen-specific, MHC-restricted, protective immune responses. After initiating the immune response, additional vaccinations may increase the protection or conversely downregulate the immune response. Groups of mice were vaccinated several times with DCs transduced with the MART-1 gene, and the anti-tumor protection was compared with that of mice receiving a single vaccination. C3H mice had poorer protection from a syngeneic MART-1-expressing tumor challenge with multiple vaccinations. This was accompanied by lower levels of splenic CTL effectors and a shift from a type 1 to a type 2 cytokine profile. On the contrary, multiple vaccinations in C57BL/6 mice generated greater in vivo antitumor protection with no decrease in splenic CTLs and no cytokine shift. Antiadenoviral humoral or cellular immune responses did not seem to contribute to these effects. When studies were performed in Fas receptor-negative C3H.(lpr) mice, the adverse effect of multiple vaccinations disappeared, and there was no cytokine shift pattern. In conclusion, C3H mice but not C57BL/6 mice receiving multiple vaccinations with DCs expressing the MART-1 tumor antigen show decreased protection associated with deviation from a type 1 to a type 2 cytokine response attributable to a Fas-receptor mediated clearance of antigen-specific IFN-gamma-producing cells.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunization Schedule , Melanoma, Experimental/immunology , fas Receptor/physiology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Interferon-gamma/immunology , MART-1 Antigen , Major Histocompatibility Complex/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation/genetics , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Tumor Cells, Cultured , Vaccination , fas Receptor/genetics
14.
J Immunother ; 23(1): 59-66, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10687138

ABSTRACT

The murine melanoma B16 expresses the murine counterpart of the human MART-1/Melan-A (MART-1) antigen, sharing a 68.6% amino acid sequence identity. In this study, mice were vaccinated with bone marrow-derived murine dendritic cells genetically modified with a replication-incompetent adenoviral vector to express the human MART-1 gene (AdVMART1). This treatment generated a protective response to a lethal tumor challenge of unmodified murine B16 melanoma cells. The response was mediated by major histocompatibility complex class I-restricted cytotoxic T lymphocytes specific for MART-1 antigen, which produced high levels of interferon-gamma when reexposed to MART-1 in vitro and lysed targets in a calcium-dependent mechanism suggestive of perforin/granzyme B lysis. MART-1 was presented by the dendritic cells used for vaccination and not by epitopes cross-presented by host antigen-presenting cells. In conclusion, dendritic cells genetically modified to express the human MART-1 antigen generate potent murine MART-1-specific protective responses to B16 melanoma.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Melanoma, Experimental/prevention & control , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Animals , Antigens, Neoplasm/genetics , Cross Reactions , Female , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , MART-1 Antigen , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Spleen/cytology , Spleen/immunology , Vaccination
15.
Mol Immunol ; 37(16): 943-50, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11395133

ABSTRACT

Human alpha-fetoprotein (AFP) is a potentially important target for the immunotherapy of hepatocellular carcinoma (HCC). AFP(542-550) (GVALQTMKQ) is one of several HLA-A2.1-restricted immunodominant AFP peptides that consistently generate AFP-specific T cell responses in human T cell cultures and in HLA-A2.1/K(b) transgenic (A2.1 tg) mice. We performed a fine specificity analysis of this nonamer to determine which amino acid side chains were critical for T cell priming and recognition. Using peptide-pulsed dendritic cells (DC) as an immunization strategy, we characterized the effects of AFP(542-550) amino acid substitutions on priming and recognition in A2.1 tg mice. Replacing the glutamine at anchor position 9 with a leucine enhanced MHC binding and AFP-specific T cell responses. Substitution of leucine at non-anchor position 4 with an alanine did not alter binding but greatly diminished T cell recognition. Computer-generated three-dimensional models provided the structural rationale for these observed effects in MHC binding and T cell responses resulted from the modifications in the AFP(542-550) sequence.


Subject(s)
HLA-A2 Antigen/immunology , Immunodominant Epitopes/immunology , T-Lymphocytes/immunology , alpha-Fetoproteins/immunology , Amino Acids/immunology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Humans , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Lymphocyte Activation , Mice , Models, Molecular , Oligopeptides/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell
16.
Cancer Gene Ther ; 6(6): 523-36, 1999.
Article in English | MEDLINE | ID: mdl-10608349

ABSTRACT

A murine model of dendritic cell (DC)-based genetic immunization to a defined human melanoma antigen (Ag), MART-1/Melan-A (MART-1), was developed. The MART-1 gene was stably transfected into the nonimmunogenic mouse fibrosarcoma cell line NFSA that is syngeneic in C3Hf/Sem/Kam (C3H, H-2k) mice to generate the NFSA(MART1) cell line. In vivo protection from a lethal NFSA(MART1) tumor challenge could be generated by DCs transduced with a recombinant adenovirus (AdV) vector expressing MART-1 (AdVMART1). This model has the following characteristics: (a) immunological specificity and memory, (b) comparable protection for varying transduction multiplicities of infection, cell doses, and sites of DC inoculation but, interestingly, worse protection with increasing numbers of vaccinations, (c) the ability to treat small established tumors, (d) an absolute requirement for CD8 and CD4 T cells, (e) generation of MART-1-specific splenic cytotoxic T lymphocytes, and (f) up-regulation of both T helper type 1 and T helper type 2 cytokines. Genetically engineered DCs presenting defined tumor Ags represent an attractive method to generate effective immune responses.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Melanoma, Experimental/therapy , Animals , Antigens, Neoplasm/genetics , Disease Models, Animal , Female , Genetic Therapy , Lung Neoplasms/secondary , Lung Neoplasms/therapy , MART-1 Antigen , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C3H , Neoplasm Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Cancer Res ; 59(17): 4369-74, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485485

ABSTRACT

An E1B gene-attenuated adenovirus (dl1520) has been proposed to have a selective cytolytic activity in cancer cells with a mutation or deletion in the p53 tumor suppressor gene (p53-null), a defect present in almost half of human hepatocellular carcinomas (HCCs). In this study, the in vitro and in vivo antitumor activity of dl1520 was investigated focusing on two human HCC cell lines, a p53-wild type (p53-wt) cell line and a p53-null cell line. dl1520 was tested for in vitro cytopathic effects and viral replication in the human HCC cell lines Hep3B (p53-null) and HepG2 (p53-wt). The in vivo antitumor effects of dl1520 were investigated in tumors grown s.c. in a severe combined immunodeficient mouse model. In addition, the combination of dl1520 infection with systemic chemotherapy was assessed in these tumor xenografts. At low multiplicities of infection, dl1520 had an apparent p53-dependent in vitro viral growth in HCC cell lines. At higher multiplicities of infection, dl1520 viral replication was independent of the p53 status of the target cells. In vivo, dl1520 significantly retarded the growth of the p53-null Hep3B xenografts, an effect augmented by the addition of cisplatin. However, complete tumor regressions were rare, and most tumors eventually grew progressively. dl1520 had no effect on the in vivo growth of the p53-wt HepG2 cells, with or without cisplatin treatment. The E1B-deleted adenoviral vector dl1520 has an apparent p53-dependent effect in HCC cell lines. However, this effect is lost at higher viral doses and only induces partial tumor regressions without tumor cures in a human HCC xenograft model.


Subject(s)
Adenoviridae/physiology , Adenovirus E1B Proteins/physiology , Carcinoma, Hepatocellular/therapy , Genes, p53/physiology , Liver Neoplasms/therapy , Virus Replication , Animals , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Cancer Res ; 59(13): 3064-7, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10397245

ABSTRACT

The majority of human hepatocellular carcinomas overexpress alpha-fetoprotein (AFP). Two genetic immunization strategies were used to determine whether AFP could serve as a target for T-cell immune responses. Dendritic cells engineered to express AFP produced potent T-cell responses in mice, as evidenced by the generation of AFP-specific CTLs, cytokine-producing T cells, and protective immunity. AFP plasmid-based immunization generated less potent responses. These novel observations demonstrate that this oncofetal antigen can serve as an effective tumor rejection antigen. This provides a rational, gene therapy-based strategy for this disease, which is responsible for the largest number of cancer-related deaths worldwide.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy , Immunotherapy , Liver Neoplasms, Experimental/therapy , Liver Neoplasms/therapy , T-Lymphocytes/immunology , alpha-Fetoproteins/genetics , Animals , Cytotoxicity, Immunologic , Female , Genetic Vectors , Humans , Liver Neoplasms/immunology , Liver Neoplasms, Experimental/immunology , Lymphoma/immunology , Major Histocompatibility Complex , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Transcription, Genetic , alpha-Fetoproteins/immunology
19.
Cancer Res ; 59(13): 3134-42, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10397256

ABSTRACT

Alpha-fetoprotein (AFP) is often derepressed in human hepatocellular carcinoma. Peptide fragments of AFP presented in the context of major histocompatibility molecules could serve as potential recognition targets by CD8 T cells, provided these lymphocytes were not clonally deleted in ontogeny. We therefore wished to determine whether the human T-cell repertoire could recognize AFP-derived peptide epitopes in the context of a common class I allele, HLA-A2.1. Dendritic cells genetically engineered to express AFP were capable of generating AFP-specific T-cell responses in autologous human lymphocyte cultures and in HLA-A2.1/Kb transgenic mice. These T cells recognize a 9-mer peptide derived from the AFP protein hAFP(542-550) (GVALQTMKQ). Identified as a potential A2.1-restricted peptide epitope from a computer analysis of the AFP sequence, hAFP(542-550) proved to have low binding affinity to A2.1, but slow off-kinetics. AFP-specific CTL- and IFN-gamma-producing cells recognize hAFP(542-550)-pulsed targets. Conversely, hAFP(542-550) peptide-generated T cells from both human lymphocyte cultures and A2.1/Kb transgenic mice recognized AFP-transfected targets in both cytotoxicity assays and cytokine release assays. These lines of evidence clearly demonstrate that AFP-reactive clones have not been deleted from the human T-cell repertoire and identify one immunodominant A2.1-restricted epitope. These findings also clearly establish AFP as a potential target for T-cell-based immunotherapy.


Subject(s)
Cytotoxicity, Immunologic , Epitopes/pharmacology , HLA-A2 Antigen/immunology , T-Lymphocytes/immunology , alpha-Fetoproteins/immunology , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Dendritic Cells/immunology , Epitopes/chemistry , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Transgenic , Recombinant Proteins/immunology , Structure-Activity Relationship , T-Lymphocytes/drug effects , Transfection , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/pharmacology
20.
Anticancer Res ; 19(2A): 1165-70, 1999.
Article in English | MEDLINE | ID: mdl-10368670

ABSTRACT

Peptides extracted from tumor cells after mild acid treatment can function as antigenic epitopes when presented by cultured dendritic cells. Peptides were extracted from four tumors syngeneic to C3H mice, three weakly immunogenic tumors (FSA, MCAK, HCA) and one non-immunogenic tumor (NFSA). Dendritic cells pulsed with peptides extracted from the three weakly immunogenic tumors partially protect mice from a tumor challenge with the parental cell line. This protection was evident by a slower rate of tumor appearance and a slower tumor growth curve when compared to control, non-immunized mice. However, vaccination of mice with dendritic cells pulsed with peptides derived from the non-immunogenic cell line NFSA did not elicit a protective response. Neither the route of immunization, the number of immunizations, nor the amount of peptides significantly affected the antitumor protection. Dendritic cells genetically engineered to produce IL-2 did not increase the protective effect of peptide-pulsed dendritic cells. These results suggest that only a partial protection against immunogenic tumors can be achieved when dendritic cells pulsed with acid-eluted tumor peptides are used as antitumor vaccination.


Subject(s)
Dendritic Cells/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/therapy , Acids , Animals , Female , Immunization , Interleukin-2/biosynthesis , Mice , Mice, Inbred C3H , Neoplasms, Experimental/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...