Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 5: 4, 2017.
Article in English | MEDLINE | ID: mdl-28116088

ABSTRACT

BACKGROUND: We previously demonstrated that tumor irradiation potentiates cancer vaccines using genetic modification of tumor cells in murine tumor models. To investigate whether tumor irradiation augments the immune response to MUC1 tumor antigen, we have tested the efficacy of tumor irradiation combined with an MVA-MUC1-IL2 cancer vaccine (Transgene TG4010) for murine renal adenocarcinoma (Renca) cells transfected with MUC1. METHODS: Established subcutaneous Renca-MUC1 tumors were treated with 8 Gy radiation on day 11 and peritumoral injections of MVA-MUC1-IL2 vector on day 12 and 17, or using a reverse sequence of vaccine followed by radiation. Growth delays were monitored by tumor measurements and histological responses were evaluated by immunohistochemistry. Specific immunity was assessed by challenge with Renca-MUC1 cells. Generation of tumor-specific T cells was detected by IFN-γ production from splenocytes stimulated in vitro with tumor lysates using ELISPOT assays. RESULTS: Tumor growth delays observed by tumor irradiation combined with MVA-MUC1-IL-2 vaccine were significantly more prolonged than those observed by vaccine, radiation, or radiation with MVA empty vector. The sequence of cancer vaccine followed by radiation two days later resulted in 55-58% complete responders and 60% mouse long-term survival. This sequence was more effective than that of radiation followed by vaccine leading to 24-30% complete responders and 30% mouse survival. Responding mice were immune to challenge with Renca-MUC1 cells, indicating the induction of specific tumor immunity. Histology studies of regressing tumors at 1 week after therapy, revealed extensive tumor destruction and a heavy infiltration of CD45+ leukocytes including F4/80+ macrophages, CD8+ cytotoxic T cells and CD4+ helper T cells. The generation of tumor-specific T cells by combined therapy was confirmed by IFN-γ secretion in tumor-stimulated splenocytes. An abscopal effect was measured by rejection of an untreated tumor on the contralateral flank to the tumor treated with radiation and vaccine. CONCLUSIONS: These findings suggest that cancer vaccine given prior to local tumor irradiation augments an immune response targeted at tumor antigens that results in specific anti-tumor immunity. These findings support further exploration of the combination of radiotherapy with cancer vaccines for the treatment of cancer.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/radiotherapy , Interferon-gamma/immunology , Interleukin-2/immunology , Mucin-1/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/radiation effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Genetic Vectors , Interferon-gamma/genetics , Interferon-gamma/therapeutic use , Interleukin-2/genetics , Interleukin-2/therapeutic use , Mice , Mucin-1/genetics , Mucin-1/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects , Vaccines, DNA , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
2.
PLoS One ; 8(9): e73310, 2013.
Article in English | MEDLINE | ID: mdl-24019914

ABSTRACT

Tumor progression is promoted by Tumor-Associated Macrophages (TAMs) and metastasis-induced bone destruction by osteoclasts. Both myeloid cell types depend on the CD115-CSF-1 pathway for their differentiation and function. We used 3 different mouse cancer models to study the effects of targeting cancer host myeloid cells with a monoclonal antibody (mAb) capable of blocking CSF-1 binding to murine CD115. In mice bearing sub-cutaneous EL4 tumors, which are CD115-negative, the anti-CD115 mAb depleted F4/80(+) CD163(+) M2-type TAMs and reduced tumor growth, resulting in prolonged survival. In the MMTV-PyMT mouse model, the spontaneous appearance of palpable mammary tumors was delayed when the anti-CD115 mAb was administered before malignant transition and tumors became palpable only after termination of the immunotherapy. When administered to mice already bearing established PyMT tumors, anti-CD115 treatment prolonged their survival and potentiated the effect of chemotherapy with Paclitaxel. As shown by immunohistochemistry, this therapeutic effect correlated with the depletion of F4/80(+)CD163(+) M2-polarized TAMs. In a breast cancer model of bone metastasis, the anti-CD115 mAb potently blocked the differentiation of osteoclasts and their bone destruction activity. This resulted in the inhibition of cancer-induced weight loss. CD115 thus represents a promising target for cancer immunotherapy, since a specific blocking antibody may not only inhibit the growth of a primary tumor through TAM depletion, but also metastasis-induced bone destruction through osteoclast inhibition.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Macrophages/immunology , Neoplasms, Experimental/therapy , Osteoclasts/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/immunology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Survival Analysis
3.
Mol Ther ; 16(5): 985-94, 2008 May.
Article in English | MEDLINE | ID: mdl-18388930

ABSTRACT

Numerous preclinical and clinical studies have shown that interleukin-2 (IL-2) induces regression of metastatic tumors. We have conducted a phase I/II, multicenter, open-label, dose-escalating study to evaluate the safety, efficacy, and biological effects of repeated intratumoral injections of adenovirus-IL-2 (TG1024) in patients with advanced solid tumors and melanoma. Thirty five patients (twenty-five with metastatic melanoma and ten with other solid tumors) were treated in eight successive cohorts at dose levels ranging from 3 x 10(8) to 3 x 10(11) viral particles (vp). Intratumoral TG1024 injections in combination with dacarbazine (DTIC) were tested in metastatic melanoma in one cohort. No clinical responses were observed at doses below 3 x 10(11) vp. Six local objective responses were recorded in patients receiving 3 x 10(11) vp per treatment [five in metastatic melanoma and one in metastatic squamous cell carcinoma (SCC) of the skin], of which two were complete responses (CRs). Most of the common side effects were injection site reactions and flu-like syndrome. TG1024 dose intensification across cohorts resulted in increased serum IL-2 levels after the injection. Intratumoral TG1024 injection induced pronounced inflammation of the treated lesion, with predominant CD8(+), TIA+ lymphocytic infiltrate. Our results show that intratumoral injections of TG1024 are safe and well tolerated. The clinical activity of TG1024 observed in this study warrants further investigations.


Subject(s)
Adenoviridae/genetics , Dacarbazine/pharmacology , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Interleukin-2/genetics , Melanoma/genetics , Melanoma/therapy , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , Female , Humans , Interleukin-2/metabolism , Male , Middle Aged , Neoplasm Metastasis
4.
Proc Natl Acad Sci U S A ; 101 Suppl 2: 14567-71, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15333750

ABSTRACT

DNA vaccines, comprised of plasmid DNA encoding proteins from pathogens, allergens, and tumors, are being evaluated as prophylactic vaccines and therapeutic treatments for infectious diseases, allergies, and cancer; plasmids encoding normal human proteins are likewise being tested as vaccines and treatments for autoimmune diseases. Examples of in vivo prophylaxis and immunotherapy, based on different types of immune responses (humoral and cellular), in a variety of disease models and under evaluation in early phase human clinical trials are presented. Viral vectors continue to show better levels of expression than those achieved by DNA plasmid vectors. We have focused our clinical efforts, at this time, on the use of recombinant viral vectors for both vaccine as well as cytokine gene transfer studies. We currently have four clinical programs in cancer immunotherapy. Two nonspecific immunotherapy programs are underway that apply adenoviral vectors for the transfer of cytokine genes into tumors in situ. An adenovirus-IFN gamma construct (TG1042) is currently being tested in phase II clinical trials in cutaneous lymphoma. A similar construct, adenovirus-IL2 (TG1024), also injected directly into solid tumors, is currently being tested in patients with solid tumors (about one-half of which are melanoma). Encouraging results are seen in both programs. Two cancer vaccine immunotherapy programs focus on two cancer-associated antigens: human papilloma virus E6 and E7 proteins and the epithelial cancer-associated antigen MUC1. Both are encoded by a highly attenuated vaccinia virus vector [modified vaccinia Ankara (MVA)] and both are coexpressed with IL-2. Encouraging results seen in both of these programs are described.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Vaccines, DNA/therapeutic use , Antigens, Neoplasm , Breast Neoplasms/therapy , Cancer Vaccines/genetics , Cytokines/genetics , Female , Genetic Therapy , Humans , Kidney Neoplasms/therapy , Lung Neoplasms/therapy , Male , Neoplasms/immunology , Papillomavirus Infections/therapy , Prostatic Neoplasms/therapy , Uterine Cervical Neoplasms/therapy , Vaccines, DNA/genetics
6.
Blood ; 104(6): 1631-8, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15161670

ABSTRACT

Primary cutaneous lymphomas have been successfully treated with interferons (IFNs), counterbalancing the T-helper 2 (Th2)-skewing state. We undertook a phase 1, open-label, dose-escalating trial of repeated intratumoral administration of TG1042 in patients with advanced primary cutaneous T-cell lymphomas (CTCLs) and multilesional cutaneous B-cell lymphomas (CBCLs). TG1042 is a third-generation, nonreplicating human adenovirus vector containing a human IFN-gamma cDNA insert. Nine patients (7 CTCL, 2 CBCL) were enrolled at the following TG1042 doses: 3 x 10(9), 3 x 10(10), and 3 x 10(11) total particles. Local clinical response was observed in 5 of 9 treated patients (3 patients with complete response [CR] and 2 patients with partial response [PR]). Out of these, 3 patients showed systemic CR with the clearance of other noninjected skin lesions. Clinical response lasted for a median of 3 months (range, 1-6 months). Adverse events were mostly of grades 1 and 2. Seven of 9 treated patients had a detectable TG1042-derived IFN-gamma message in injected lesions after the first treatment cycle. A TG1042-IFN-gamma message was also detectable after several treatment cycles. We demonstrate the induction of humoral immune response to lymphoma tumor-antigen se70-2 after treatment. Our study shows that intralesional injections of TG1042 are both safe and well tolerated.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Immunotherapy/methods , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphoma/therapy , Skin Neoplasms/therapy , Adenoviridae/physiology , Adult , Aged , Aged, 80 and over , Female , Gene Expression , Humans , Immunohistochemistry , Interferon-gamma/adverse effects , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
7.
Cancer Gene Ther ; 11(1): 61-72, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14681727

ABSTRACT

To circumvent the toxicity caused by systemic injection of cytokines, cytokine cDNA genes encoding the human interleukin IL-2 cDNA (Ad-IL-2) and murine interferon IFN-gamma gene (Ad- IFN-gamma) were inserted into adenoviral vectors. These constructs were used for intratumoral gene therapy of murine renal adenocarcinoma Renca tumors. Treatment with three doses of Ad-IL-2 or Ad- IFN-gamma, given a day apart, was more effective than single-dose gene therapy. We found that tumor irradiation enhanced the therapeutic efficacy of Ad-IL-2 and Ad-IFN-gamma intratumoral gene therapy. Tumor irradiation, administered 1 day prior to three doses of Ad-IL-2 treatment, was more effective than radiation or Ad-IL-2 alone, resulting in tumor growth arrest in all mice, increased survival and a consistent increase in complete tumor regression response rate. Complete responders rejected Renca tumor challenge and demonstrated specific cytotoxic T-cell activity, indicative of specific tumor immunity. The effect of radiation combined with three doses of Ad-IFN-gamma was less pronounced and did not lead to tumor immunity. Histological observations showed that irradiation of the tumor prior to gene therapy increased tumor destruction and inflammatory infiltrates in the tumor nodules. These findings demonstrate that tumor irradiation improves the efficacy of Ad-IL-2 gene therapy for induction of antitumor immune response.


Subject(s)
Genetic Therapy/methods , Interferon-gamma/genetics , Interferon-gamma/therapeutic use , Interleukin-2/genetics , Interleukin-2/therapeutic use , Kidney Neoplasms/radiotherapy , Kidney Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Flow Cytometry , Gene Expression Regulation, Neoplastic , Genetic Vectors/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/adverse effects , Interferon-gamma/metabolism , Interleukin-2/adverse effects , Interleukin-2/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mice , Neoplasm Transplantation , Survival Rate , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transgenes/genetics , Up-Regulation
8.
Microbiology (Reading) ; 143 ( Pt 8): 2733-2741, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9274026

ABSTRACT

Four Lactobacillus strains (Lb. plantarum NCIMB 8826, Lb. paracasei LbTGS1.4, Lb. casei ATCC 393 and Lb. fermentum KLD) were tested for their ability to produce and secrete heterologous proteins. These strains were first screened with an alpha-amylase reporter under the control of a set of expression or expression/secretion signals from various lactic acid bacteria. With most of the constructions tested, the level of extracellular production was highest in Lb. plantarum NCIMB 8826, and lowest in Lb. paracasei LbTGS1.4. These two strains were next assayed using a model antigen consisting of the N-terminal part of the M6 protein from Streptococcus pyogenes fused to the linear epitope ELDKWAS from human immunodeficiency virus gp41 protein. Secretion of this heterologous protein was inefficient in Lb. paracasei LbTGS1.4, which accumulated a large intracellular pool of the unprocessed precursor, whereas Lb. plantarum NCIMB 8826 was able to secrete the antigen to a level as high as 10 mg l-1.


Subject(s)
Antigens, Bacterial , Antigens/metabolism , Bacterial Outer Membrane Proteins , Bacterial Proteins/metabolism , Carrier Proteins , HIV Envelope Protein gp41/metabolism , Lactobacillus/genetics , Protein Engineering/methods , Antigens/genetics , Bacterial Proteins/genetics , Biological Transport , Genes, Reporter , Genetic Vectors , HIV Envelope Protein gp41/genetics , Lactobacillus/metabolism , Plasmids , Proteins/metabolism , Recombinant Proteins/metabolism , Transformation, Genetic , alpha-Amylases/genetics , alpha-Amylases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...