Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother ; 36(8): 442-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23994887

ABSTRACT

We created a vaccine in which irradiated allogeneic lung adenocarcinoma cells are combined with a bystander K562 cell line transfected with hCD40L and hGM-CSF. By recruiting and activating dendritic cells, we hypothesized that the vaccine would induce tumor regression in metastatic lung adenocarcinoma. Intradermal vaccine was given q14 days×3, followed by monthly ×3. Cyclophosphamide (300 mg/m IV) was administered before the first and fourth vaccines to deplete regulatory T cells. All-trans retinoic acid was given (150/mg/m/d) after the first and fourth vaccines to enhance dendritic cell differentiation. Twenty-four participants were accrued at a single institution from October 2006 to June 2008, with a median age 64 years and median of 4 previous lines of systemic therapy. A total of 101 vaccines were administered. Common toxicities were headache (54%) and site reaction (38%). No radiologic responses were observed. Median overall survival was 7.9 months and median progression-free survival was 1.7 months. Of 14 patients evaluable for immunological study, 5 had peptide-induced CD8 T-cell activation after vaccination. Overall, vaccine administration was feasible in an extensively pretreated population of metastatic lung cancer. Despite a suggestion of clinical activity in the subset with immune response, the trial did not meet the primary endpoint of inducing radiologic tumor regression.


Subject(s)
Adenocarcinoma/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Dendritic Cells/immunology , Lung Neoplasms/therapy , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Aged , Antigen Presentation , Antigens, Neoplasm/immunology , Bystander Effect , CD40 Ligand/genetics , CD40 Ligand/metabolism , Cell Differentiation/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , K562 Cells , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lymphocyte Activation , Male , Middle Aged , Neoplasm Metastasis , Recurrence , Survival Analysis , Transgenes/genetics , Treatment Outcome , Tretinoin/administration & dosage
2.
Anticancer Res ; 31(10): 3193-204, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21965726

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is the most aggressive form of lung cancer with poor disease outcome. The chemotherapeutic agent paclitaxel (PA) is commonly used as a second-line treatment in SCLC, but response rates are low. MATERIALS AND METHODS: 86M1 SCLC cells were treated in the presence or absence of paclitaxel and TRAIL or the combination for 24 hours. Western blot analysis was utilized to examine protein expression, cell surface protein expression and membrane integrity were elucidated by flow cytometry, and immunofluorescence microscopy was used to demonstrate translocation of proteins to the cell nucleus. RESULTS: Human 86M1 SCLC cells were found to be resistant to PA killing in vitro. This resistance is mediated by up-regulation of pro-survival protein BCL-xl. However, PA also increases surface expression of death receptors 4 and 5 (DR4 and DR5, respectively). The death receptors' ligand increased SCLC killing by PA through an apparent caspase-independent route involving activation/translocation of AIF. CONCLUSION: The addition of TRAIL to PA can potentiate apoptosis in a relatively PA-resistant SCLC line (specifically 86M1 cells). More importantly, we are the first to report an active method of resistance to paclitaxel in SCLC via BCL-xl up-regulation.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/pathology , Paclitaxel/pharmacology , Small Cell Lung Carcinoma/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Receptors, Death Domain/metabolism , Up-Regulation/drug effects , bcl-X Protein/metabolism
3.
J Clin Invest ; 120(4): 1111-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20234093

ABSTRACT

Cancer immunotherapy faces a serious challenge because of low clinical efficacy. Recently, a number of clinical studies have reported the serendipitous finding of high rates of objective clinical response when cancer vaccines are combined with chemotherapy in patients with different types of cancers. However, the mechanism of this phenomenon remains unclear. Here, we tested in mice several cancer vaccines and an adoptive T cell transfer approach to cancer immunotherapy in combination with several widely used chemotherapeutic drugs. We found that chemotherapy made tumor cells more susceptible to the cytotoxic effect of CTLs through a dramatic perforin-independent increase in permeability to GrzB released by the CTLs. This effect was mediated via upregulation of mannose-6-phosphate receptors on the surface of tumor cells and was observed in mouse and human cells. When combined with chemotherapy, CTLs raised against specific antigens were able to induce apoptosis in neighboring tumor cells that did not express those antigens. These data suggest that small numbers of CTLs could mediate a potent antitumor effect when combined with chemotherapy. In addition, these results provide a strong rationale for combining these modalities for the treatment of patients with advanced cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic/drug effects , Immunotherapy , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cisplatin/pharmacology , Dendritic Cells/immunology , Female , Granzymes/physiology , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Paclitaxel/pharmacology , Vaccination
4.
J Immunother ; 31(1): 72-80, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157014

ABSTRACT

We report a single center phase II trial of sequential vaccination followed with vaccine plus interleukin-2 (IL-2). Vaccination consisted of autologous cells cultured from primary tumor or resected metastasis, transduced to express B7.1 surface molecule and then irradiated. The vaccine would hypothetically costimulate tumor-reactive T cells before IL-2 exposure. Treatment plan was 3 subcutaneous vaccine injections at 4-week intervals and subcutaneous IL-2 treatment for 6 weeks starting at week 7. Sixty-six patients enrolled, of whom 39 received at least 1 vaccine; most observed toxicity was attributable to IL-2 not vaccine; best responses were 3% pathologic complete response, 5% partial response, 64% stable disease, and 28% disease progression. Median survival was 21.8 months (95% confidence interval 17.8 to 29.6). Significant postvaccination increases in IFN-gamma responses to autologous tumor were observed in 2/26 cases. Eighty-one percent of posttreatment subdermal delayed-type hypersensitivity tests (using nontransduced, irradiated autologous tumor cells) had biopsies demonstrating injection site lymphocytic infiltration. Post hoc comparison of the median survival of subjects whose biopsies had lymphocytic infiltration appears longer than in the 19% noninfiltrated (28.4 vs. 17.8 mo, P=0.045, two-sided log-rank test). The single arm design precludes conclusive comparison of objective response rates (not different here) or median survival (longer here) versus those of historical series using similar IL-2 schedules alone. Better outcomes could be logically associated to vaccine response (detectable lymphocytic infiltrates) or to random events that a single arm study design cannot address. This vaccine approach may merit further clinical development.


Subject(s)
B7-1 Antigen/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Renal Cell/therapy , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Adult , Aged , Aged, 80 and over , B7-1 Antigen/genetics , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Female , Humans , Interferon-gamma/metabolism , Interleukin-2/adverse effects , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Middle Aged , Neoplasm Staging , Skin/drug effects , Skin/immunology , Skin/pathology , Survival Analysis , Transfection , Treatment Outcome , Tumor Cells, Cultured
5.
J Surg Res ; 125(2): 173-81, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15854671

ABSTRACT

BACKGROUND: Tumors evade T cell-mediated rejection despite the presence of tumor associated antigens (TAAs) and T cells specific for these TAAs in cancer patients. Therapeutic tumor vaccines are being developed to prevent this evasion. Previous reports revealed that anti-tumor T cell responses could be activated in mice when granulocyte macrophage-colony stimulating factor (GM-CSF) or CD40L are produced at tumor vaccine sites. We sought to test the hypothesis that production of GM-CSF and CD40L by a bystander cell line could induce an anti-tumor T cell response in an in vitro human model. MATERIALS AND METHODS: The K562 cell line was stably transfected with the human GM-CSF and CD40L genes. The effect of this cell line on T cell responses was tested in a human autologous mixed tumor cell/lymph node cell model using tissue from a series of cancer patients. RESULTS: There was no significant anti-tumor T cell response when human lymphocytes derived from tumor-draining lymph nodes were stimulated with autologous tumor cells in vitro. However, significant anti-tumor T cell responses were observed when bystander cells transfected with CD40L and GM-CSF were added to the cultures. CONCLUSIONS: A fully autologous human model consisting of tumor cells as stimulator cells and tumor-draining lymph nodes as responder cells can be used to test immunotherapeutic strategies. T cells in these lymph nodes are unresponsive to autologous tumor cells, but this lack of responsiveness can be reversed in the presence of GM-CSF and CD40L. These data provide a rationale for testing tumor cell vaccines incorporating GM-CSF- and CD40L-expressing bystanders in clinical trials.


Subject(s)
CD40 Ligand/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , K562 Cells/metabolism , Lymph Nodes/metabolism , T-Lymphocytes/metabolism , Bystander Effect , Cancer Vaccines , Humans , Immunotherapy , In Vitro Techniques , Lymphocyte Activation , Neoplasms/metabolism , Neoplasms/therapy , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...