Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Res ; 67(13): 6451-8, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616706

ABSTRACT

We have previously shown preferential tumor-homing and therapeutic efficacy of adoptively transferred type 1 CTL (Tc1) when compared with type 2 CTL (Tc2) in mice bearing intracranial ovalbumin-transfected melanoma (M05). Further characterizing the expression of a panel of homing receptors on Tc1 and Tc2 cells, we found that very late antigen (VLA)-4 (a heterodimer of CD49d and CD29), but none of other receptors evaluated, was expressed at significantly higher levels on Tc1 cells than on Tc2 cells. Although CD49d (alpha(4) integrin) can form heterodimers with both beta(1) (CD29) and beta(7) integrins, alpha(4)beta(7) complexes were not expressed by either Tc1 or Tc2 cells, suggesting that CD49d is solely expressed in VLA-4 complexes. VLA-4 expression on Tc2 cells was down-regulated in an interleukin (IL)-4 dose-dependent manner but not by other type 2 cytokines, such as IL-10 and IL-13, suggesting that IL-4 uniquely down-regulates VLA-4 expression on these cells. In accordance with the differential expression of VLA-4 on Tc1 versus Tc2 cells, Tc1 cells alone were competent to adhere to plate-bound VCAM-1-Ig fusion protein. Finally, the efficient trafficking of Tc1 cells into intracranial M05 lesions in vivo was efficiently blocked by administration of monoclonal antibodies against CD49d or VCAM-1 or small interfering RNA-mediated silencing of CD49d on Tc1 cells. Collectively, these data support the critical role of VLA-4 in the effective intracranial tumor homing of adoptive-transferred, antigen-specific Tc1 cells and suggest that more effective vaccine and/or ex vivo T-cell activation regimens may be developed by promoting the generation of VLA-4(+) antitumor Tc1 cells.


Subject(s)
Central Nervous System Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Integrin alpha4beta1/physiology , T-Lymphocytes, Cytotoxic/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/chemistry , Integrin alpha4/biosynthesis , Integrin alpha4beta1/metabolism , Integrin beta1/biosynthesis , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Ovalbumin/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
2.
J Transl Med ; 5: 10, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17295916

ABSTRACT

BACKGROUND: Toll-like receptor (TLR)3 ligands serve as natural inducers of pro-inflammatory cytokines capable of promoting Type-1 adaptive immunity, and TLR3 is abundantly expressed by cells within the central nervous system (CNS). To improve the efficacy of vaccine strategies directed against CNS tumors, we evaluated whether administration of a TLR3 ligand, polyinosinic-polycytidylic (poly-IC) stabilized with poly-lysine and carboxymethylcellulose (poly-ICLC) would enhance the anti-CNS tumor effectiveness of tumor peptide-based vaccinations. METHODS: C57BL/6 mice bearing syngeneic CNS GL261 glioma or M05 melanoma received subcutaneous (s.c.) vaccinations with synthetic peptides encoding CTL epitopes--mEphA2 (671-679), hgp100 (25-33) and mTRP-2 (180-188) for GL261, or ovalbumin (OVA: 257-264) for M05. The mice also received intramuscular (i.m.) injections with poly-ICLC. RESULTS: The combination of subcutaneous (s.c.) peptide-based vaccination and i.m. poly-ICLC administration promoted systemic induction of antigen (Ag)-specific Type-1 CTLs expressing very late activation antigen (VLA)-4, which confers efficient CNS-tumor homing of vaccine-induced CTLs based on experiments with monoclonal antibody (mAb)-mediated blockade of VLA-4. In addition, the combination treatment allowed expression of IFN-gamma by CNS tumor-infiltrating CTLs, and improved the survival of tumor bearing mice in the absence of detectable autoimmunity. CONCLUSION: These data suggest that poly-ICLC, which has been previously evaluated in clinical trials, can be effectively combined with tumor Ag-specific vaccine strategies, thereby providing a greater index of therapeutic efficacy.


Subject(s)
Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Epitopes/immunology , Glioma/prevention & control , Peptides/immunology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Toll-Like Receptor 3/immunology , Vaccination , Animals , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/pharmacology , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Ephrin-A2/immunology , Glioma/immunology , Glioma/therapy , Humans , Injections, Intramuscular , Integrin alpha4beta1/immunology , Ligands , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Poly I-C/administration & dosage , Polylysine/administration & dosage , Polylysine/pharmacology , Staining and Labeling , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Up-Regulation/drug effects
3.
Cancer Res ; 66(11): 5883-91, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740728

ABSTRACT

Restricted and high-level expression of interleukin-13 receptor alpha2 (IL-13Ralpha2) in a majority of human malignant gliomas makes this protein an attractive vaccine target. We have previously described the identification of the IL-13Ralpha2(345-353) peptide as a human leukocyte antigen-A2 (HLA-A2)-restricted CTL epitope. However, as it remains unclear how efficiently peptide-based vaccines can induce specific CTLs in patients with malignant gliomas, we have examined whether analogue epitopes could elicit heteroclitic antitumor T-cell responses versus wild-type peptides. We have created three IL-13Ralpha2 analogue peptides by substitutions of the COOH-terminal isoleucine (I) for valine (V) and the NH(2)-terminal tryptophan (W) for either alanine (A), glutamic acid (E), or nonsubstituted (W; designated as 1A9V, 1E9V, and 9V, respectively). In comparison with the native IL-13Ralpha2 epitope, the analogue peptides 9V and 1A9V displayed higher levels of binding affinity and stability in HLA-A2 complexes and yielded an improved stimulatory index for patient-derived, specific CTLs against the native epitope expressed by HLA-A2(+) glioma cells. In HLA-A2-transgenic HHD mice, immunization with the peptides 9V and 1A9V induced enhanced levels of CTL reactivity and protective immunity against an intracranial challenge with IL13Ralpha2-expressing syngeneic tumors when compared with vaccines containing the native IL-13Ralpha2 epitope. These findings indicate highly immunogenic IL-13Ralpha2 peptide analogues may be useful for the development of vaccines capable of effectively expanding IL-13Ralpha2-specific, tumor-reactive CTLs in glioma patients.


Subject(s)
Cancer Vaccines/immunology , Glioma/immunology , Glioma/therapy , Peptide Fragments/immunology , Receptors, Interleukin/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , HLA-A2 Antigen/immunology , Humans , Interleukin-13 Receptor alpha1 Subunit , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Peptide Fragments/pharmacology , Receptors, Interleukin-13 , T-Lymphocytes, Cytotoxic/drug effects
4.
Cancer Res ; 66(8): 4478-87, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618775

ABSTRACT

The development of effective immunotherapeutic strategies for central nervous system (CNS) tumors requires a firm understanding of factors regulating the trafficking of tumor antigen-specific CTLs into CNS tumor lesions. Using C57BL/6 mice bearing intracranial (i.c.) ovalbumin-transfected melanoma (M05), we evaluated the efficacy and tumor homing of i.v. transferred type 1 or 2 CTLs (Tc1 or Tc2, respectively) prepared from ovalbumin-specific T-cell receptor-transgenic OT-1 mice. We also tested our hypothesis that intratumoral (i.t.) delivery of dendritic cells that had been transduced with IFN-alpha cDNA (DC-IFN-alpha) would enhance the tumor-homing and antitumor effectiveness of adoptively transferred Tc1 via induction of an IFN-gamma-inducible protein 10 (IP-10). In vitro, DC-IFN-alpha induced IP-10 production by M05 and enhanced the cytolytic activity of Tc1. In vivo, i.v. transferred Tc1 trafficked efficiently into i.c. M05 and mediated antitumor responses more effectively than Tc2, and their effect was IP-10 dependent. I.t. injections of DC-IFN-alpha remarkably enhanced the tumor homing, therapeutic efficacy, and in situ IFN-gamma production of i.v. delivered Tc1, resulting in the long-term survival and persistence of systemic ovalbumin-specific immunity. These data suggest that Tc1-based adoptive transfer therapy may represent an effective modality for CNS tumors, particularly when combined with strategies that promote a type 1 polarized tumor microenvironment.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , Chemokines, CXC/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain Neoplasms/genetics , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Dendritic Cells/immunology , Immunologic Memory , Interferon-alpha/genetics , Interferon-alpha/immunology , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Transfection
5.
Neoplasia ; 7(8): 717-22, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16207473

ABSTRACT

EphA2 is a receptor tyrosine kinase and is frequently overexpressed in a wide array of advanced cancers. We demonstrate in the current study that the EphA2 protein is restrictedly expressed in primary glioblastoma multiforme and anaplastic astrocytoma tissues in comparison to normal brain tissues. To evaluate the possibility of targeting EphA2 in glioma vaccine strategies, we stimulated human leukocyte antigen (HLA) A2+ peripheral blood mononuclear cells (PBMCs) obtained from healthy donors and glioma patients with autologous dendritic cells (DCs) loaded with synthetic EphA2883-891 peptide (TLADFDPRV), which has previously been reported to induce interferon-gamma in HLA-A2+ PBMCs. Stimulated PBMCs demonstrated antigen-specific cytotoxic T lymphocyte (CTL) responses as detected by specific lysis of T2 cells loaded with the EphA2883 peptide as well as HLA-A2+ glioma cells, SNB19 and U251, that express EphA2. Furthermore, in vivo immunization of HLA-A2 transgenic HHD mice with the EphA2883-891 peptide resulted in the development of an epitope-specific CTL response in splenocytes, despite the fact that EphA2883-891 is an autoantigen in these mice. Taken together, these data suggest that EphA2883-891 may be an attractive antigen epitope for molecularly targeted glioma vaccines.


Subject(s)
Antigens, Neoplasm/biosynthesis , Brain Neoplasms/immunology , Cancer Vaccines/pharmacology , Glioblastoma/immunology , Receptor, EphA2/biosynthesis , Animals , Antigens, Neoplasm/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , HLA-A2 Antigen/immunology , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Transgenic , Receptor, EphA2/administration & dosage , Receptor, EphA2/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology
6.
J Immunol ; 175(4): 2730-40, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16081851

ABSTRACT

We tested whether modulation of the CNS-tumor microenvironment by delivery of IFN-alpha-transduced dendritic cells (DCs: DC-IFN-alpha) would enhance the therapeutic efficacy of peripheral vaccinations with cytokine-gene transduced tumor cells. Mice bearing intracranial GL261 glioma or MCA205 sarcoma received peripheral immunizations with corresponding irradiated tumor cells engineered to express IL-4 or GM-CSFs, respectively, as well as intratumoral delivery of DC-IFN-alpha. This regimen prolonged survival of the animals and induced tumor-specific CTLs that expressed TRAIL, which in concert with perforin and Fas ligand (FasL) was involved in the tumor-specific CTL activity of these cells. The in vivo antitumor activity associated with this approach was abrogated by administration of neutralizing mAbs against TRAIL or FasL and was not observed in perforin-/-, IFN-gamma-/-, or FasL-/- mice. Transduction of the tumor cells with antiapoptotic protein cellular FLIP rendered the gene-modified cells resistant to TRAIL- or FasL-mediated apoptosis and to CTL killing activity in vitro. Furthermore, the combination therapeutic regimen was ineffective in an intracranial cellular FLIP-transduced MCA205 brain tumor model. These results suggest that the combination of intratumoral delivery of DC-IFN-alpha and peripheral immunization with cytokine-gene transduced tumor cells may be an effective therapy for brain tumors that are sensitive to apoptotic signaling pathways.


Subject(s)
Apoptosis/immunology , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Glioma/immunology , Interferon-alpha/administration & dosage , Signal Transduction/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Adoptive Transfer , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Cytokines/biosynthesis , Cytokines/physiology , Cytokines/therapeutic use , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Glioma/pathology , Glioma/prevention & control , Injections, Intralesional , Injections, Intraventricular , Interferon-alpha/genetics , Interferon-alpha/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Transduction, Genetic
7.
J Immunol ; 174(11): 7194-201, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905564

ABSTRACT

We previously demonstrated that IL-4 gene-transfected glioma cell vaccines induce effective therapeutic immunity in preclinical glioma models, and have initiated phase I trials of these vaccines in patients with malignant gliomas. To gain additional mechanistic insight into the efficacy of this approach, we have treated mice bearing the MCA205 (H-2(b)) or CMS-4 (H-2(d)) sarcomas. IL-12/23 p40(-/-) and IFN-gamma(-/-) mice, which were able to reject the initial inoculation of IL-4 expressing tumors, failed to mount a sustained systemic response against parental (nontransfected) tumor cells. Paracrine production of IL-4 in vaccine sites promoted the accumulation and maturation of IL-12p70-secreting tumor-infiltrating dendritic cells (TIDCs). Adoptive transfer of TIDCs isolated from vaccinated wild-type, but not IL-12/23 p40(-/-), mice were capable of promoting tumor-specific CTL responses in syngeneic recipient animals. Interestingly, both STAT4(-/-) and STAT6(-/-) mice failed to reject IL-4-transfected tumors in concert with the reduced capacity of TIDCs to produce IL-12p70 and to promote specific antitumor CTL reactivity. These results suggest that vaccines consisting of tumor cells engineered to produce the type 2 cytokine, IL-4, critically depend on type 1 immunity for their observed therapeutic efficacy.


Subject(s)
Cancer Vaccines/immunology , Cell Movement/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Fibrosarcoma/immunology , Interleukin-4/genetics , Sarcoma, Experimental/immunology , Transfection , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Cell Movement/genetics , DNA-Binding Proteins/physiology , Dendritic Cells/pathology , Female , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Graft Rejection/genetics , Graft Rejection/immunology , Immunity, Cellular/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/physiology , Interleukin-12/administration & dosage , Interleukin-12/genetics , Interleukin-12/physiology , Interleukin-12 Subunit p40 , Interleukin-4/administration & dosage , Interleukin-4/biosynthesis , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Protein Subunits/physiology , STAT4 Transcription Factor , STAT6 Transcription Factor , Sarcoma, Experimental/genetics , Sarcoma, Experimental/pathology , T-Lymphocyte Subsets/immunology , Trans-Activators/physiology
8.
Cancer Gene Ther ; 12(9): 757-68, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15832173

ABSTRACT

We have created a novel cellular vehicle for gene therapy of malignant gliomas by transfection of murine bone marrow stroma cells (MSCs) with a cDNA encoding epidermal growth factor receptor (EGFR). These cells (EGFR-MSCs) demonstrate enhanced migratory responses toward glioma-conditioned media in comparison to primary MSCs in vitro. Enhanced migration of EGFR-MSC was at least partially dependent on EGF-EGFR, PI3-, MAP kinase kinase, and MAP kinases, protein kinase C, and actin polymerization. Unlike primary MSCs, EGFR-MSCs were resistant to FasL-mediated cytotoxicity and were capable of stimulating allogeneic mixed lymphocyte reaction, suggesting EGFR-MSCs possess suitable characteristics as vehicles for brain tumor immuno-gene therapy. Following injection at various sites, including the contralateral hemisphere in the brain of syngeneic mice, EGFR-MSCs were able to migrate toward GL261 gliomas or B16 melanoma in vivo. Finally, intratumoral injection with EGFR-MSC adenovirally engineered to secrete interferon-alpha to intracranial GL261 resulted in significantly prolonged survival in comparison to controls. These data indicate that EGFR-MSCs may serve as attractive vehicles for infiltrating brain malignancies such as malignant gliomas.


Subject(s)
Bone Marrow Cells/physiology , Brain Neoplasms/therapy , Cell Movement , ErbB Receptors/genetics , Genetic Therapy/methods , Glioma/therapy , Actins/metabolism , Animals , Bone Marrow Cells/cytology , Cell Line , Epidermal Growth Factor/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Stromal Cells/physiology , Transfection
9.
J Transl Med ; 2(1): 40, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15563374

ABSTRACT

BACKGROUND: A novel tyrosine kinase receptor EphA2 is expressed at high levels in advanced and metastatic cancers. We examined whether vaccinations with synthetic mouse EphA2 (mEphA2)-derived peptides that serve as T cell epitopes could induce protective and therapeutic anti-tumor immunity. METHODS: C57BL/6 mice received subcutaneous (s.c.) vaccinations with bone marrow-derived dendritic cells (DCs) pulsed with synthetic peptides recognized by CD8+ (mEphA2671-679, mEphA2682-689) and CD4+ (mEphA230-44) T cells. Splenocytes (SPCs) were harvested from primed mice to assess the induction of cytotoxic T lymphocyte (CTL) responses against syngeneic glioma, sarcoma and melanoma cell lines. The ability of these vaccines to prevent or treat tumor (s.c. injected MCA205 sarcoma or B16 melanoma; i.v. injected B16-BL6) establishment/progression was then assessed. RESULTS: Immunization of C57BL/6 mice with mEphA2-derived peptides induced specific CTL responses in SPCs. Vaccination with mEPhA2 peptides, but not control ovalbumin (OVA) peptides, prevented the establishment or prevented the growth of EphA2+ or EphA2-negative syngeneic tumors in both s.c. and lung metastasis models. CONCLUSIONS: These data indicate that mEphA2 can serve as an attractive target against which to direct anti-tumor immunity. The ability of mEphA2 vaccines to impact EphA2-negative tumors such as the B16 melanoma may suggest that such beneficial immunity may be directed against alternative EphA2+ target cells, such as the tumor-associated vascular endothelial cells.

10.
Cancer Res ; 64(16): 5830-8, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313927

ABSTRACT

We evaluated the effects, on immunity and survival, of injection of interferon (IFN)-alpha-transfected dendritic cells (DC-IFN-alpha) into intracranial tumors in mice immunized previously with syngeneic dendritic cells (DCs) pulsed either with ovalbumin-derived CTL or T helper epitopes. These immunizations protected animals from s.c. challenge with ovalbumin-expressing M05 melanoma (class I+ and class II-negative). Notably, antiovalbumin CTL responses were observed in animals vaccinated with an ovalbumin-derived T helper epitope but only after the mice were challenged with M05 cells. This cross-priming of CTL was dependent on both CD4+ and CD8+ T cells. Because we observed that s.c., but not intracranial, tumors were infiltrated with CD11c+ DCs, and because IFN-alpha promotes the activation and survival of both DCs and T cells, we evaluated the combinational antitumor effects of injecting adenoviral (Ad)-IFN-alpha-engineered DCs into intracranial M05 tumors in preimmunized mice. Delivery of DC-IFN-alpha prolonged survival. This was most notable for animals prevaccinated with both the CTL and T helper ovalbumin epitopes, with 60% (6 of 10) of mice (versus 0 of 10 of control animals) surviving for > 80 days after tumor challenge. DC-IFN-alpha appeared to persist longer than mock-transfected DCs within the intracranial tumor microenvironment, and DC-IFN-alpha-treated mice exhibited enhanced levels of ovalbumin-specific CTL in draining cervical lymph nodes. On the basis of these results, we believe that local expression of IFN-alpha by DCs within the intracranial tumor site may enhance the clinical efficacy of peripheral vaccine approaches for brain tumors.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Interferon-alpha/immunology , Amino Acid Sequence , Animals , Brain Neoplasms/immunology , Brain Neoplasms/prevention & control , Epitopes, T-Lymphocyte/immunology , Female , Interferon-alpha/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/prevention & control , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Ovalbumin/immunology , Ovalbumin/pharmacology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...