Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 23(15): 4046-4054, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28280092

ABSTRACT

Purpose: Several lines of evidence support targeting the androgen signaling pathway in breast cancer. Enzalutamide is a potent inhibitor of androgen receptor signaling. Preclinical data in estrogen-expressing breast cancer models demonstrated activity of enzalutamide monotherapy and enhanced activity when combined with various endocrine therapies (ET). Enzalutamide is a strong cytochrome P450 3A4 (CYP3A4) inducer, and ETs are commonly metabolized by CYP3A4. The pharmacokinetic (PK) interactions, safety, and tolerability of enzalutamide monotherapy and in combination with ETs were assessed in this phase I/Ib study.Experimental Design: Enzalutamide monotherapy was assessed in dose-escalation and dose-expansion cohorts of patients with advanced breast cancer. Additional cohorts examined effects of enzalutamide on anastrozole, exemestane, and fulvestrant PK in patients with estrogen receptor-positive/progesterone receptor-positive (ER+/PgR+) breast cancer.Results: Enzalutamide monotherapy (n = 29) or in combination with ETs (n = 70) was generally well tolerated. Enzalutamide PK in women was similar to prior data on PK in men with prostate cancer. Enzalutamide decreased plasma exposure to anastrozole by approximately 90% and exemestane by approximately 50%. Enzalutamide did not significantly affect fulvestrant PK. Exposure of exemestane 50 mg/day given with enzalutamide was similar to exemestane 25 mg/day alone.Conclusions: These results support a 160 mg/day enzalutamide dose in women with breast cancer. Enzalutamide can be given in combination with fulvestrant without dose modifications. Exemestane should be doubled from 25 mg/day to 50 mg/day when given in combination with enzalutamide; this combination is being investigated in a randomized phase II study in patients with ER+/PgR+ breast cancer. Clin Cancer Res; 23(15); 4046-54. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Phenylthiohydantoin/analogs & derivatives , Adult , Aged , Anastrozole , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/adverse effects , Benzamides , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytochrome P-450 CYP3A/genetics , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/classification , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Middle Aged , Neoplasm Staging , Nitriles/administration & dosage , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/adverse effects , Phenylthiohydantoin/pharmacokinetics , Postmenopause , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Triazoles/administration & dosage
2.
Clin Cancer Res ; 22(15): 3774-81, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26858312

ABSTRACT

PURPOSE: Preclinical evidence suggests that both docetaxel and enzalutamide target androgen receptor translocation and signaling. This phase Ib study assessed the safety, tolerability, and pharmacokinetics of docetaxel when administered with enzalutamide as first-line systemic chemotherapy in men with metastatic castration-resistant prostate cancer (mCRPC). EXPERIMENTAL METHODS: Docetaxel-naïve patients received 21-day cycles of docetaxel (75 mg/m(2)). Enzalutamide (160 mg/day) was administered daily starting on day 2 of cycle 1. Patients were allowed to stop and restart docetaxel at any time following cycle 2. Treatment continued indefinitely until unacceptable toxicity or discontinuation due to investigator or patient preference. RESULTS: A total of 22 patients received docetaxel, of whom 21 also received enzalutamide. Docetaxel was administered for a median of 5.0 cycles and enzalutamide for a median of 12.0 months. With concomitant treatment, geometric mean docetaxel exposure decreased by 11.8%, whereas peak concentrations decreased by 3.7% relative to docetaxel alone. The most common toxicities observed during the period of concomitant therapy were neutropenia (86.4%) and fatigue (77.3%). Common toxicities observed with post-docetaxel enzalutamide were constipation (23.8%), decreased appetite (19.0%), fatigue (19.0%), and musculoskeletal pain (19.0%). Treatment with enzalutamide and docetaxel resulted in prostate-specific antigen decreases in almost all patients based on exploratory analysis of available baseline and on-study prostate-specific antigen data. CONCLUSIONS: The combination of docetaxel and enzalutamide is feasible, although higher rates of neutropenia and neutropenic fever than anticipated were observed. Reductions in docetaxel exposure with enzalutamide coadministration were not considered clinically meaningful. This combination warrants further study in a larger mCRPC population. Clin Cancer Res; 22(15); 3774-81. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides , Combined Modality Therapy , Docetaxel , Drug Monitoring , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/mortality , Retreatment , Taxoids/administration & dosage , Treatment Outcome
3.
J Clin Oncol ; 31(32): 4105-14, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-24101053

ABSTRACT

PURPOSE: Increased hepatocyte growth factor/MET signaling is associated with poor prognosis and acquired resistance to epidermal growth factor receptor (EGFR) -targeted drugs in patients with non-small-cell lung cancer (NSCLC). We investigated whether dual inhibition of MET/EGFR results in clinical benefit in patients with NSCLC. PATIENTS AND METHODS: Patients with recurrent NSCLC were randomly assigned at a ratio of one to one to receive onartuzumab plus erlotinib or placebo plus erlotinib; crossover was allowed at progression. Tumor tissue was required to assess MET status by immunohistochemistry (IHC). Coprimary end points were progression-free survival (PFS) in the intent-to-treat (ITT) and MET-positive (MET IHC diagnostic positive) populations; additional end points included overall survival (OS), objective response rate, and safety. RESULTS: There was no improvement in PFS or OS in the ITT population (n = 137; PFS hazard ratio [HR], 1.09; P = .69; OS HR, 0.80; P = .34). MET-positive patients (n = 66) treated with erlotinib plus onartuzumab showed improvement in both PFS (HR, .53; P = .04) and OS (HR, .37; P = .002). Conversely, clinical outcomes were worse in MET-negative patients treated with onartuzumab plus erlotinib (n = 62; PFS HR, 1.82; P = .05; OS HR, 1.78; P = .16). MET-positive control patients had worse outcomes versus MET-negative control patients (n = 62; PFS HR, 1.71; P = .06; OS HR, 2.61; P = .004). Incidence of peripheral edema was increased in onartuzumab-treated patients. CONCLUSION: Onartuzumab plus erlotinib was associated with improved PFS and OS in the MET-positive population. These results combined with the worse outcomes observed in MET-negative patients treated with onartuzumab highlight the importance of diagnostic testing in drug development.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cross-Over Studies , Disease-Free Survival , Double-Blind Method , Erlotinib Hydrochloride , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Proportional Hazards Models , Proto-Oncogene Proteins c-met/biosynthesis , Quinazolines/administration & dosage , Quinazolines/adverse effects , Treatment Outcome
4.
J Clin Oncol ; 30(1): 34-41, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22124101

ABSTRACT

PURPOSE: Metastatic melanoma, a highly vascularized tumor with strong expression of vascular endothelial growth factor, has an overall poor prognosis. We conducted a placebo-controlled, double-blind phase II study of carboplatin plus paclitaxel with or without bevacizumab in patients with previously untreated metastatic melanoma. PATIENTS AND METHODS: Patients were randomly assigned in a two-to-one ratio to carboplatin (area under the curve, 5) plus paclitaxel (175 mg/m(2)) and bevacizumab (15 mg/kg; CPB) or placebo (CP) administered intravenously once every 3 weeks. Progression-free survival (PFS) was the primary end point. Secondary end points included overall survival (OS) and safety. RESULTS: Two hundred fourteen patients (73% with M1c disease) were randomly assigned. With a median follow-up of 13 months, median PFS was 4.2 months for the CP arm (n = 71) and 5.6 months for the CPB arm (n = 143; hazard ratio [HR], 0.78; P = .1414). Overall response rates were 16.4% and 25.5%, respectively (P = .1577). With 13-month follow-up, median OS was 8.6 months in the CP arm versus 12.3 months in the CPB arm (HR, 0.67; P = .0366), whereas in an evaluation 4 months later, it was 9.2 versus 12.3 months, respectively (HR, 0.79; P = .1916). In patients with elevated serum lactate dehydrogenase (n = 84), median PFS and OS were longer in the CPB arm (PFS: 4.4 v 2.7 months; HR, 0.62; OS: 8.5 v 7.5 months; HR, 0.52). No new safety signals were observed. CONCLUSION: The study did not meet the primary objective of statistically significant improvement in PFS with the addition of bevacizumab to carboplatin plus paclitaxel. A larger phase III study will be necessary to determine whether there is benefit to the addition of bevacizumab to carboplatin plus paclitaxel in this disease setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Carboplatin/administration & dosage , Double-Blind Method , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Male , Melanoma/pathology , Middle Aged , Paclitaxel/administration & dosage , Skin Neoplasms/pathology , Treatment Outcome
5.
Cancer Res ; 69(7): 3077-85, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293190

ABSTRACT

Despite the frequent detection of circulating tumor antigen-specific T cells, either spontaneously or following active immunization or adoptive transfer, immune-mediated cancer regression occurs only in the minority of patients. One theoretical rate-limiting step is whether effector T cells successfully migrate into metastatic tumor sites. Affymetrix gene expression profiling done on a series of metastatic melanoma biopsies revealed a major segregation of samples based on the presence or absence of T-cell-associated transcripts. The presence of lymphocytes correlated with the expression of defined chemokine genes. A subset of six chemokines (CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10) was confirmed by protein array and/or quantitative reverse transcription-PCR to be preferentially expressed in tumors that contained T cells. Corresponding chemokine receptors were found to be up-regulated on human CD8(+) effector T cells, and transwell migration assays confirmed the ability of each of these chemokines to promote migration of CD8(+) effector cells in vitro. Screening by chemokine protein array identified a subset of melanoma cell lines that produced a similar broad array of chemokines. These melanoma cells more effectively recruited human CD8(+) effector T cells when implanted as xenografts in nonobese diabetic/severe combined immunodeficient mice in vivo. Chemokine blockade with specific antibodies inhibited migration of CD8(+) T cells. Our results suggest that lack of critical chemokines in a subset of melanoma metastases may limit the migration of activated T cells, which in turn could limit the effectiveness of antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines/biosynthesis , Melanoma/immunology , Melanoma/metabolism , Cell Line, Tumor , Cell Movement/immunology , Chemokines/genetics , Chemokines/immunology , Gene Expression , Humans , Melanoma/genetics , Multigene Family , Protein Array Analysis
6.
Prostate ; 69(2): 142-8, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18942640

ABSTRACT

BACKGROUND: Increasing evidence suggests that prostate cancer is visible to the immune system and is potentially responsive to immunotherapeutic interventions. Previous work has identified prostate-specific membrane antigen (PSMA) as a potential antigen for T-cell recognition, and specific PSMA epitopes presented by HLA-A2 have been described. One vaccination strategy that is being pursued in the clinic utilizes peptide-pulsed peripheral blood mononuclear cells (PBMC) + IL-12. METHODS: HLA-A2(+) patients with castrate-resistant prostate cancer and normal organ function were considered. Vaccines were prepared using autologous PBMC loaded with a PSMA peptide previously reported to be immunogenic (LLHETDSAV) administered subcutaneously every 3 weeks. T-cell responses and tumor activity were assessed. RESULTS: Although the vaccine was well tolerated, no clinical responses were observed in 12 enrolled patients and no immune responses were detected based on an ex vivo IFN-gamma ELISPOT assay. To examine immunogenicity of this PSMA peptide in more detail, an in vitro priming assay was utilized with normal donor PBMC, and no detectable reactivity was observed. CONCLUSIONS: Our results suggest that the PSMA peptide LLHETDSAV is poorly immunogenic in humans and that alternative prostate cancer antigens should be pursued.


Subject(s)
Adenocarcinoma/immunology , Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Peptide Fragments/immunology , Prostatic Neoplasms/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Cancer Vaccines/therapeutic use , Epitopes/immunology , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/therapeutic use , Male , Neoplasm Metastasis/immunology , Patient Selection , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Recombinant Proteins/therapeutic use , T-Lymphocytes/immunology
7.
Nat Immunol ; 7(11): 1166-73, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17028589

ABSTRACT

T cell anergy has been correlated with defective signaling by the GTPase Ras, but causal and mechanistic data linking defective Ras activity with T cell anergy are lacking. Here we used adenoviral transduction to genetically manipulate nonproliferating T cells and show that active Ras restored interleukin 2 production and mitogen-activated protein kinase signaling in T cells that were made anergic in vitro or in vivo. Diacylglycerol kinases (DGKs), which negatively regulate Ras activity, were upregulated in anergic T cells, and a DGK inhibitor restored interleukin 2 production in anergic T cells. Both anergy and DGK-alpha overexpression were associated with defective translocation of the Ras guanine nucleotide-exchange factor RasGRP1 to the plasma membrane. Our data support a causal function for excess DGK activity and defective Ras signaling in T cell anergy.


Subject(s)
Clonal Anergy/immunology , Diacylglycerol Kinase/physiology , Th1 Cells/enzymology , Th1 Cells/immunology , ras Proteins/physiology , Animals , Cell Line , Immunophenotyping , Isoenzymes/physiology , Mice , Mice, Transgenic
8.
Invest New Drugs ; 24(2): 141-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16514482

ABSTRACT

PURPOSE: Bryostatin-1 is a PKC modulator with direct anti-tumor activity and immunomodulatory properties. We combined different doses of Bryostatin-1 with IL-2 to determine effects on clinical response rate and T cell phenotype in patients with advanced kidney cancer. EXPERIMENTAL DESIGN: IL-2 naïve patients were given 11 x 10(6) IU subcutaneously of IL-2 on days 1-4, 8-11, and 15-18 of every 28-day cycle. Twenty four patients were randomized to treatment cohorts of 5, 15 or 25 mcg/m2 of Bryostatin-1 on days 1, 8 and 15, starting in the second cycle. An additional nine, non-randomized patients were given 35 mcg/m2. Lymphocytes were analyzed for number, activation status, and production of IL-2, IL-4 and IFN-gamma. Response evaluation was performed every 3 cycles. RESULTS: Common grade 3 toxicities included fatigue (5), nausea/vomiting (5), myopathy (3), dyspnea (3), and syncope (3). Four patients, in the two highest dose cohorts, demonstrated evidence of tumor shrinkage, although there was only 1 objective PR. The median time to progression was 104 days (95% CI 88-120) and the median survival was 452 days (95% CI = 424-480). There was no significant boosting effect of Bryostatin-1 on lymphocytes. CONCLUSIONS: The addition of Bryostatin-1 to IL-2 was well tolerated, but the overall response rate was low (3.2%), indicating that further studies with this combination are not warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bryostatins/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interleukin-2/administration & dosage , Kidney Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bryostatins/adverse effects , Carcinoma, Renal Cell/immunology , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Female , Humans , Interleukin-2/adverse effects , Kidney Neoplasms/immunology , Male , Middle Aged , Neoplasm Metastasis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Treatment Failure
9.
Cancer Immunol Immunother ; 55(10): 1185-97, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16468035

ABSTRACT

Although melanoma tumors usually express antigens that can be recognized by T cells, immune-mediated tumor rejection is rare. In many cases this is despite the presence of high frequencies of circulating tumor antigen-specific T cells, suggesting that tumor resistance downstream from T cell priming represents a critical barrier. Analyzing T cells directly from the melanoma tumor microenvironment, as well as the nature of the microenvironment itself, is central for understanding the key downstream mechanisms of tumor escape. In the current report we have studied tumor-associated lymphocytes from a patient with metastatic melanoma and large volume malignant ascites. The ascites fluid showed abundant tumor cells that expressed common melanoma antigens and retained expression of class I MHC and antigen processing machinery. The ascites fluid contained the chemokines CCL10, CCL15, and CCL18 which was associated with a large influx of activated T cells, including CD8(+) T cells recognizing HLA-A2 tetramer complexes with peptides from Melan-A and NA17-A. However, several functional defects of these tumor antigen-specific T cells were seen, including poor production of IFN-gamma in response to peptide-pulsed APC or autologous tumor cells, and lack of expression of perforin. Although these defects were T cell intrinsic, we also observed abundant CD4(+)CD25(+)FoxP3(+) T cells, as well as transcripts for FoxP3, IL-10, PD-L1/B7-H1, and indoleamine-2,3-dioxygenase (IDO). Our observations suggest that, despite recruitment of large numbers of activated CD8(+) T cells into the tumor microenvironment, T cell hyporesponsiveness and additional negative regulatory mechanisms can limit the effector phase of the anti-tumor immune response.


Subject(s)
Ascites/immunology , Ascitic Fluid/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/immunology , Adult , Antigen Presentation/immunology , Antigens, CD , Antigens, Differentiation/immunology , Antigens, Differentiation/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Ascites/etiology , Ascitic Fluid/chemistry , Ascitic Fluid/cytology , CTLA-4 Antigen , Cancer Vaccines , Chemokines/immunology , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/therapeutic use , Immunotherapy , Interleukin-2/immunology , Interleukin-2/therapeutic use , Lymphocyte Activation/immunology , Male , Melanoma/complications , Melanoma/therapy , Membrane Glycoproteins/immunology , Membrane Glycoproteins/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , Tumor Escape , gp100 Melanoma Antigen
10.
Clin Cancer Res ; 10(12 Pt 1): 4048-54, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15217937

ABSTRACT

PURPOSE: Vascular endothelial growth factor (VEGF) expression is prognostic in melanoma, and the activity of VEGF is mediated in part through the receptor tyrosine kinase Flk-1. A Phase II study of SU5416, a preferential inhibitor of Flk-1, was carried out in patients with metastatic melanoma to determine clinical response, tolerability, and changes in tumor vascular perfusion. EXPERIMENTAL DESIGN: Patients with documented progressive disease and

Subject(s)
Indoles/therapeutic use , Melanoma/drug therapy , Pyrroles/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adult , Aged , Angiogenesis Inhibitors/therapeutic use , Female , Glucose/metabolism , Humans , Lymphocytes/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Metastasis , Neovascularization, Pathologic , Protein Kinase Inhibitors/therapeutic use , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism
11.
Cancer Res ; 64(3): 1140-5, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871849

ABSTRACT

Although increased circulating tumor antigen-specific CD8(+) T cells can be achieved by vaccination or adoptive transfer, tumor progression nonetheless often occurs through resistance to effector function. To develop a model for identifying mechanisms of resistance to antigen-specific CTLs, poorly immunogenic B16-F10 melanoma was transduced to express the K(b)-binding peptide SIYRYYGL as a green fluorescent protein fusion protein that should be recognized by high-affinity 2C TCR transgenic T cells. Although B16.SIY cells expressed high levels of antigen and were induced to express K(b) in response to IFN-gamma, they were poorly recognized by primed 2C/RAG2(-/-) T cells. A screen for candidate inhibitory ligands revealed elevated PD-L1/B7H-1 on IFN-gamma-treated B16-F10 cells and also on eight additional mouse tumors and seven human melanoma cell lines. Primed 2C/RAG2(-/-)/PD-1(-/-) T cells showed augmented cytokine production, proliferation, and cytolytic activity against tumor cells compared with wild-type 2C cells. This effect was reproduced with anti-PD-L1 antibody present during the effector phase but not during the priming culture. Adoptive transfer of 2C/RAG2(-/-)/PD-1(-/-) T cells in vivo caused tumor rejection under conditions in which wild-type 2C cells or CTLA-4-deficient 2C cells did not reject. Our results support interfering with PD-L1/PD-1 interactions to augment the effector function of tumor antigen-specific CD8(+) T cells in the tumor microenvironment.


Subject(s)
B7-1 Antigen , Blood Proteins/physiology , CD8-Positive T-Lymphocytes/immunology , Neoplasms, Experimental/immunology , Peptides/physiology , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD , Antigens, Differentiation/immunology , B7-H1 Antigen , Blood Proteins/antagonists & inhibitors , Blood Proteins/biosynthesis , Blood Proteins/genetics , CTLA-4 Antigen , Interferon-gamma/pharmacology , Membrane Glycoproteins , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Peptides/antagonists & inhibitors , Peptides/genetics , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics , Transduction, Genetic , Up-Regulation/drug effects
12.
Eur J Immunol ; 33(10): 2687-95, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14515252

ABSTRACT

The adapter protein CrkL has been implicated in multiple signal transduction pathways in hematopoietic cells. In T lymphocytes, the recruitment of CrkL-C3G complexes has been correlated with hyporesponsiveness, implicating CrkL as a potential negative regulator. To test this hypothesis we examined T cell activation in CrkL-deficient mice. The CrkL(-/-) genotype was partially embryonic lethal. In viable CrkL(-/-) mice, peripheral blood counts were normal. The thymus from CrkL(-/-) mice had 40% fewer cells compared to littermates, but the proportion of thymocyte subsets was comparable. There was no discernable alteration in T cell function as reflected by T cell numbers, expression of memory markers, IL-2 production, proliferation, and differentiation into Th1/Th2 phenotypes. Immunization induced comparable levels of IgG2a and IgG1 antibodies. Chimeric mice, generated by transfer of CrkL(-/-) fetal liver cells into irradiated RAG2(-/-) recipients, also showed normal T cell function, arguing against selection via partial embryonic lethality. Our results indicate that CrkL is not absolutely required for T cell development or function, and argue against it being an essential component of a negative regulatory pathway in TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Nuclear Proteins/physiology , T-Lymphocytes/physiology , Animals , Antibody Formation , Cell Differentiation , DNA-Binding Proteins/physiology , Interferon-gamma/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Th1 Cells/physiology , Th2 Cells/physiology
13.
J Clin Oncol ; 21(12): 2342-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12805336

ABSTRACT

PURPOSE: Preclinical studies showed that immunization with peripheral blood mononuclear cells (PBMC) loaded with tumor antigen peptides plus interleukin-12 (IL-12) induced CD8+ T-cell responses and tumor rejection. We recently determined that recombinant human (rh) IL-12 at 30 to 100 ng/kg is effective as a vaccine adjuvant in patients. A phase II study of immunization with Melan-A peptide-pulsed PBMC + rhIL-12 was conducted in 20 patients with advanced melanoma. PATIENTS AND METHODS: Patients were HLA-A2-positive and had documented Melan-A expression. Immunization was performed every 3 weeks with clinical re-evaluation every three cycles. Immune responses were measured by ELISpot assay before and after treatment and through the first three cycles, and were correlated with clinical outcome. RESULTS: Most patients had received prior therapy and had visceral metastases. Nonetheless, two patients achieved a complete response, five patients achieved a minor or mixed response, and four patients had stable disease. The median survival was 12.25 months for all patients and was not yet reached for those with a normal lactate dehydrogenase. There were no grade 3 or 4 toxicities. Measurement of specific CD8+ T-cell responses by direct ex vivo ELISpot revealed a significant increase in interferon gamma-producing T cells against Melan-A (P =.015) after vaccination, but not against an Epstein-Barr virus control peptide (P =.86). There was a correlation between the magnitude of the increase in Melan-A-specific cells and clinical response (P =.046). CONCLUSION: This immunization approach may be more straightforward than dendritic cell strategies and seems to have clinical activity that can be correlated to a biologic end point.


Subject(s)
Interleukin-12/metabolism , Leukocytes, Mononuclear/metabolism , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Neoplasm Proteins/therapeutic use , Recombinant Proteins/metabolism , Adult , Aged , Antigens, Neoplasm , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines , Female , HLA-A2 Antigen/biosynthesis , Humans , Interferon-gamma , MART-1 Antigen , Male , Middle Aged , Neoplasm Metastasis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...