Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 9(12): 1451-1464, 2021 12.
Article in English | MEDLINE | ID: mdl-34635485

ABSTRACT

Immune-checkpoint blockade has revolutionized cancer treatment. However, most patients do not respond to single-agent therapy. Combining checkpoint inhibitors with other immune-stimulating agents increases both efficacy and toxicity due to systemic T-cell activation. Protease-activatable antibody prodrugs, known as Probody therapeutics (Pb-Tx), localize antibody activity by attenuating capacity to bind antigen until protease activation in the tumor microenvironment. Herein, we show that systemic administration of anti-programmed cell death ligand 1 (anti-PD-L1) and anti-programmed cell death protein 1 (anti-PD-1) Pb-Tx to tumor-bearing mice elicited antitumor activity similar to that of traditional PD-1/PD-L1-targeted antibodies. Pb-Tx exhibited reduced systemic activity and an improved nonclinical safety profile, with markedly reduced target occupancy on peripheral T cells and reduced incidence of early-onset autoimmune diabetes in nonobese diabetic mice. Our results confirm that localized PD-1/PD-L1 inhibition by Pb-Tx can elicit robust antitumor immunity and minimize systemic immune-mediated toxicity. These data provide further preclinical rationale to support the ongoing development of the anti-PD-L1 Pb-Tx CX-072, which is currently in clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/therapeutic use , Immunotherapy/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Tumor Microenvironment
2.
Biochem Cell Biol ; 97(5): 638-646, 2019 10.
Article in English | MEDLINE | ID: mdl-30986357

ABSTRACT

We recently demonstrated that Cav1 (caveolin-1) is a negative regulator of Stat3 (signal transducer and activator of transcription-3) activity in mouse fibroblasts and human lung carcinoma SHP77 cells. We now examined whether the cellular context may affect their levels as well as the relationship between them, by assessing Cav1 and Stat3-ptyr705 amounts in different cell lines. In MDA-MB-231, A549, and HaCat cells, Cav1 levels were high and Stat3-ptyr705 levels were low, consistent with the notion of a negative effect of endogenous Cav1 on Stat3-ptyr705 levels in these lines. In addition, manipulation of Cav1 levels revealed a negative effect in MCF7 and mouse fibroblast cells, while Cav1 upregulation induced apoptosis in MCF7 cells. In contrast, however, line MRC9 had high Cav1 and high Stat3-ptyr705 levels, indicating that high Cav1 is insufficient to reduce Stat3-ptyr705 levels in this line. MCF7 and LuCi6 cells had very low Cav1 and Stat3-ptyr705 levels, indicating that the low Stat3-ptyr705 can be independent from Cav1 levels altogether. Our results reveal a further level of complexity in the relationship between Cav1 and Stat3-ptyr705 than previously thought. In addition, we demonstrate that in a feedback loop, Stat3 inhibition upregulates Cav1 in HeLa cells but not in other lines tested.


Subject(s)
Breast Neoplasms/metabolism , Caveolin 1/metabolism , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Tyrosine/metabolism , Animals , Caveolin 1/antagonists & inhibitors , Cells, Cultured , Female , Humans , Mice , Mice, Inbred BALB C
3.
Cancer Res ; 78(17): 4943-4956, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29945961

ABSTRACT

Currently approved inhibitors of the PD-1/PD-L1 pathway represent a major advance for the treatment of lung cancers, yet they are ineffective in a majority of patients due to lack of preexisting T-cell reactivity. Here, we show that a TLR9 agonist delivered by inhalation is able to prime T-cell responses against poorly immunogenic lung tumors and to complement the effects of PD-1 blockade. Inhaled TLR9 agonist causes profound remodeling in tumor-bearing lungs, leading to the formation of tertiary lymphoid structures adjacent to the tumors, CD8+ T-cell infiltration into the tumors, dendritic cell expansion, and antibody production. Inhalation of TLR9 agonist also increased the pool of functional PD-1lowT-bethigh effector CD8+ T cells in tumor-bearing lungs. Effector CD8+ T cells generated by inhaled TLR9 agonist treatment were licensed by PD-1 blockade to become highly functional CTLs, leading to a durable rejection of both lung tumors and tumor lesions outside the lungs. CD4+ T cells activated in response to inhaled TLR9 play a critical role in this process by controlling the proliferation, preventing exhaustion, and guiding the differentiation of optimally functional CTLs. This study characterizes a strategy to apply localized TLR9 stimulation to a tumor type not accessible for direct injection, a strategy that may expand the therapeutic potential of PD-1 blockade in non-small cell lung cancer.Significance: These findings demonstrate that local delivery of a toll-like receptor 9 agonist can change the immune content of an entire organ and enhance the efficacy of immune checkpoint inhibition.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/17/4943/F1.large.jpg Cancer Res; 78(17); 4943-56. ©2018 AACR.


Subject(s)
Antibodies/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Oligodeoxyribonucleotides/administration & dosage , Toll-Like Receptor 9/genetics , Administration, Inhalation , Animals , Antibodies/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Disease Models, Animal , Flow Cytometry , Humans , Mice , Oligodeoxyribonucleotides/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
PLoS One ; 9(5): e96318, 2014.
Article in English | MEDLINE | ID: mdl-24806510

ABSTRACT

Cellular microenvironments, particularly those found in tumors, elicit a tolerogenic DC phenotype which can attenuate immune responses. Central to this process is the STAT3-mediated signaling cascade. As a transcription factor and oncogene, STAT3 promotes the expression of genes which allow tumor cells to proliferate, migrate and evade apoptosis. More importantly, activation of STAT3 in tumor infiltrating immune cells has been shown to be responsible, in part, for their immune-suppressed phenotype. The ability of STAT3 to orchestrate a diverse set of immunosuppressive instructions has made it an attractive target for cancer vaccines. Using a conditional hematopoietic knockout mouse model of STAT3, we evaluated the impact of STAT3 gene ablation on the differentiation of dendritic cells from bone marrow precursors. We also assessed the impact of STAT3 deletion on phagocytosis, maturation, cytokine secretion and antigen presentation by GM-CSF derived DCs in vitro. In addition to in vitro studies, we compared the therapeutic efficacy of DC vaccination using STAT3 deficient DCs to wild type counterparts in an intracranial mouse model of GBM. Our results indicated the following pleiotropic functions of STAT3: hematopoietic cells which lacked STAT3 were unresponsive to Flt3L and failed to differentiate as DCs. In contrast, STAT3 was not required for GM-CSF induced DC differentiation as both wild type and STAT3 null bone marrow cells gave rise to similar number of DCs. STAT3 also appeared to regulate the response of GM-CSF derived DCs to CpG. STAT3 null DCs expressed high levels of MHC-II, secreted more IL-12p70, IL-10, and TNFα were better antigen presenters in vitro. Although STAT3 deficient DCs displayed an enhanced activated phenotype in culture, they elicited comparable therapeutic efficacy in vivo compared to their wild type counterparts when utilized in vaccination paradigms in mice bearing intracranial glioma tumors.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/cytology , Dendritic Cells/physiology , Phagocytosis/physiology , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation/genetics , Glioblastoma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunotherapy , Mice , Mice, Knockout , STAT3 Transcription Factor/genetics
5.
Expert Opin Biol Ther ; 14(9): 1241-57, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24773178

ABSTRACT

INTRODUCTION: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and it carries a dismal prognosis. Adenoviral vector (Ad)-mediated gene transfer is being developed as a promising therapeutic strategy for GBM. Preclinical studies have demonstrated safety and efficacy of adenovirus administration into the brain and tumor mass in rodents and into the non-human primates' brain. Importantly, Ads have been safely administered within the tumor resection cavity in humans. AREAS COVERED: This review gives background on GBM and Ads; we describe gene therapy strategies for GBM and discuss the value of combination approaches. Finally, we discuss the results of the human clinical trials for GBM that have used Ads. EXPERT OPINION: The transduction characteristics of Ads, and their safety profile, added to their capacity to achieve high levels of transgene expression have made them powerful vectors for the treatment of GBM. Recent gene therapy successes in the treatment of retinal diseases and systemic brain metabolic diseases encourage the development of gene therapy for malignant glioma. Exciting clinical trials are currently recruiting patients; although, it is the large randomized Phase III controlled clinical trials that will provide the final decision on the success of gene therapy for the treatment of GBM.


Subject(s)
Adenoviridae , Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors , Glioma/therapy , Animals , Brain/immunology , Brain/metabolism , Glioblastoma/therapy , Humans , Immunomodulation/genetics , Transgenes
6.
J Pharmacol Exp Ther ; 349(3): 458-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696041

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) has been implicated as a hub for multiple oncogenic pathways. The constitutive activation of STAT3 is present in several cancers, including gliomas (GBMs), and is associated with poor therapeutic responses. Phosphorylation of STAT3 triggers its dimerization and nuclear transport, where it promotes the transcription of genes that stimulate tumor growth. In light of this role, inhibitors of the STAT3 pathway are attractive therapeutic targets for cancer. To this end, we evaluated the STAT3-inhibitory activities of three compounds (CPA-7 [trichloronitritodiammineplatinum(IV)], WP1066 [(S,E)-3-(6-bromopyridin-2-yl)-2-cyano-N-(1-phenylethyl)acrylamide, C17H14BrN3O], and ML116 [4-benzyl-1-{thieno[2,3-d]pyrimidin-4-yl}piperidine, C18H19N3S]) in cultured rodent and human glioma cells, including GBM cancer stem cells. Our results demonstrate a potent induction of growth arrest in GBM cells after drug treatment with a concomitant induction of cell death. Although these compounds were effective at inhibiting STAT3 phosphorylation, they also displayed variable dose-dependent inhibition of STAT1, STAT5, and nuclear factor κ light-chain enhancer of activated B cells. The therapeutic efficacy of these compounds was further evaluated in peripheral and intracranial mouse tumor models. Whereas CPA-7 elicited regression of peripheral tumors, both melanoma and GBM, its efficacy was not evident when the tumors were implanted within the brain. Our data suggest poor permeability of this compound to tumors located within the central nervous system. WP1066 and ML116 exhibited poor in vivo efficacy. In summary, CPA-7 constitutes a powerful anticancer agent in models of peripheral solid cancers. Our data strongly support further development of CPA-7-derived compounds with increased permeability to enhance their efficacy in primary and metastatic brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/secondary , STAT3 Transcription Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorine Compounds/pharmacokinetics , Chlorine Compounds/pharmacology , Chlorine Compounds/therapeutic use , Drug Screening Assays, Antitumor , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Piperidines/pharmacokinetics , Piperidines/pharmacology , Piperidines/therapeutic use , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/therapeutic use , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , STAT3 Transcription Factor/genetics , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/therapeutic use , Tissue Distribution , Tyrphostins/pharmacokinetics , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
7.
Neurotherapeutics ; 11(3): 623-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752661

ABSTRACT

In human gliomas, the RTK/RAS/PI(3)K signaling pathway is nearly always altered. We present a model of experimental gliomagenesis that elucidates the contributions of genes involved in this pathway (PDGF-B ligand, HRAS-G12V, and AKT). We also examine the effect on gliomagenesis by the potential modifier gene, IDH1-R132H. Injections of lentiviral-encoded oncogenes induce de novo gliomas of varying penetrance, tumor progression, and histological grade depending on the specific oncogenes used. Our model mimics hallmark histological structures of high-grade glioma, such as pseudopalisades, glomeruloid microvascular proliferation, and diffuse tumor invasion. We use our model of gliomagenesis to test the efficacy of an experimental brain tumor gene therapy. Our model allowed us to test the contributions of oncogenes in the RTK/RAS/PI(3)K pathway, and their potential modification by over-expression of mutated IDH1, in glioma development and progression in rats. Our model constitutes a clinically relevant system to study gliomagenesis, the effects of modifier genes, and the efficacy of experimental therapeutics.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Disease Models, Animal , Genetic Therapy , Glioma/mortality , Glioma/therapy , Humans , Lentivirus/genetics , Mutation , Rats , Rats, Sprague-Dawley , Signal Transduction , Survival Analysis
8.
Neurosci Lett ; 527(2): 71-7, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22906921

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and deadliest of adult primary brain tumors. Due to its invasive nature and sensitive location, complete resection remains virtually impossible. The resistance of GBM against chemotherapy and radiotherapy necessitate the development of novel therapies. Gene therapy is proposed for the treatment of brain tumors and has demonstrated pre-clinical efficacy in animal models. Here we review the various experimental therapies that have been developed for GBM including both cytotoxic and immune stimulatory approaches. We also review the combined conditional cytotoxic immune stimulatory therapy that our lab has developed which is dependent on the adenovirus mediated expression of the conditional cytotoxic gene, Herpes Simplex Type 1 Thymidine Kinase (TK) and the powerful DC growth factor Fms-like tyrosine kinase 3 ligand (Flt3L). Combined delivery of these vectors elicits tumor cell death and an anti-tumor adaptive immune response that requires TLR2 activation. The implications of our studies indicate that the combined cytotoxic and immunotherapeutic strategies are effective strategies to combat deadly brain tumors and warrant their implementation in human Phase I clinical trials for GBM.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/therapy , Adaptive Immunity , Animals , Brain/immunology , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Clinical Trials as Topic , Cytotoxins/genetics , Genetic Vectors , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Tumor Escape/immunology , Tumor Microenvironment
9.
Neuromethods ; 77: 229-243, 2012.
Article in English | MEDLINE | ID: mdl-31462854

ABSTRACT

Modeling human disease in small animals has been fundamental in advancing our scientific knowledge and for the development of novel therapeutic strategies. In the case of brain cancer, implantable tumor models, both intracranial and also in the periphery, have been widely used and extensively characterized. These models can be used to better understand certain aspects of tumor biology such as growth, neovascularization, response to potential therapies, and interaction with the immune system. Brain tumors from patients as well as rodents have been cultured in vitro, in an attempt to establish permanent cell lines. Human glioma tumors have also been maintained by serial passage in the flanks of immune-deficient animals, as it has been shown that it is not feasible to continuously passage them in culture. In this chapter, we describe various gliomas that have been isolated from mice, rats, and humans and subsequently used as syngeneic or xenograft tumor models in vivo. The majority of the models that we present in this chapter arose either spontaneously or by administration of chemical carcinogens. We compare and contrast the histopathological, genetic, and invasive features of the tumor lines as well as identify novel treatment modalities that have been developed. Finally, we present the procedures for intracranial implantation of tumor cells in rodents using stereotactic surgical techniques. The use of this technique enables the generation of large numbers of animals harboring intracranial tumors with relative ease and the survival of tumor-bearing animals is highly reproducible. These characteristics make the use of these in vivo models very attractive when aiming to develop and test the effectiveness of novel anticancer therapies.

10.
Neoplasia ; 13(10): 947-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028620

ABSTRACT

We have demonstrated that modifying the tumor microenvironment through intratumoral administration of adenoviral vectors (Ad) encoding the conditional cytotoxic molecule, i.e., HSV1-TK and the immune-stimulatory cytokine, i.e., fms-like tyrosine kinase 3 ligand (Flt3L) leads to T-cell-dependent tumor regression in rodent models of glioblastoma. We investigated the role of B cells during immune-mediated glioblastoma multiforme regression. Although treatment with Ad-TK+Ad-Flt3L induced tumor regression in 60% of wild-type (WT) mice, it completely failed in B-cell-deficient Igh6(-/-) mice. Tumor-specific T-cell precursors were detected in Ad-TK+Ad-Flt3L-treated WT mice but not in Igh6(-/-) mice. The treatment also failed in WT mice depleted of total B cells or marginal zone B cells. Because we could not detect circulating antibodies against tumor cells and the treatment was equally efficient in WT mice and in mice with B-cell-specific deletion of Prdm 1 (encoding Blimp-1), in which B cells are present but unable to fully differentiate into antibody-secreting plasma cells, tumor regression in this model is not dependent on B cells' production of tumor antigen-specific immunoglobulins. Instead, B cells seem to play a role as antigen-presenting cells (APCs). Treatment with Ad-TK+Ad-Flt3L led to an increase in the number of B cells in the cervical lymph nodes, which stimulated the proliferation of syngeneic T cells and induced clonal expansion of antitumor T cells. Our data show that B cells act as APCs, playing a critical role in clonal expansion of tumor antigen-specific T cells and brain tumor regression.


Subject(s)
B-Lymphocytes/immunology , Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/therapy , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cytotoxicity, Immunologic/immunology , Female , Glioblastoma/genetics , Glioblastoma/pathology , Herpesvirus 1, Human/enzymology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes/metabolism , Thymidine Kinase/genetics , Thymidine Kinase/immunology , Thymidine Kinase/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
11.
Anticancer Agents Med Chem ; 11(8): 729-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21707497

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. GBM is very aggressive due to its poor cellular differentiation and invasiveness, which makes complete surgical resection virtually impossible. Therefore, GBM's invasive nature as well as its intrinsic resistance to current treatment modalities makes it a unique therapeutic challenge. Extensive examination of human GBM specimens has uncovered that these tumors overexpress a variety of receptors that are virtually absent in the surrounding non-neoplastic brain. Human GBMs overexpress receptors for cytokines, growth factors, ephrins, urokinase-type plasminogen activator (uPA), and transferrin, which can be targeted with high specificity by linking their ligands with highly cytotoxic molecules, such as Diptheria toxin and Pseudomonas exotoxin A. We review the preclinical development and clinical translation of targeted toxins for GBM. In view of the clinical experience, we conclude that although these are very promising therapeutic modalities for GBM patients, efforts should be focused on improving the delivery systems utilized in order to achieve better distribution of the immuno-toxins in the tumor/resection cavity. Delivery of targeted toxins using viral vectors would also benefit enormously from improved strategies for local delivery.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Immunotoxins/administration & dosage , Immunotoxins/metabolism , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Humans
12.
Curr Gene Ther ; 11(3): 155-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21453286

ABSTRACT

The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioma/therapy , Toxins, Biological/therapeutic use , Animals , Brain Neoplasms/genetics , Combined Modality Therapy , Gene Targeting , Glioma/genetics , Humans , Immunotherapy , Interferons/therapeutic use , Interleukins/therapeutic use , Models, Biological
13.
Discov Med ; 10(53): 293-304, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21034670

ABSTRACT

Glioblastoma multiforme (GBM) is a deadly primary brain tumor in adults, with a median survival of ~12-18 months post-diagnosis. Despite recent advances in conventional therapeutic approaches, only modest improvements in median survival have been achieved; GBM usually recurs within 12 months post-resection, with poor prognosis. Thus, novel therapeutic strategies to target and kill GBM cells are desperately needed. Our group and others are pursuing virotherapy and gene therapy strategies for the treatment of GBM. In this review, we will discuss various virotherapy and gene therapy approaches for GBM currently under pre-clinical and clinical evaluation including direct or conditional cytotoxic, and/or immunostimulatory approaches. We also discuss cutting-edge technologies for drug/gene delivery and targeting brain tumors, including the use of stem cells as delivery platforms, the use of targeted immunotoxins, and the therapeutic potential of using GBM microvesicles to deliver therapeutic siRNAs or virotherapies. Finally, various animal models available to test novel GBM therapies are discussed.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/therapy , Oncolytic Virotherapy/methods , Therapies, Investigational/methods , Adult , Animals , Genetic Therapy/trends , Humans , Models, Biological , Oncolytic Virotherapy/trends , Therapies, Investigational/trends
14.
PLoS Med ; 6(1): e10, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19143470

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor that carries a 5-y survival rate of 5%. Attempts at eliciting a clinically relevant anti-GBM immune response in brain tumor patients have met with limited success, which is due to brain immune privilege, tumor immune evasion, and a paucity of dendritic cells (DCs) within the central nervous system. Herein we uncovered a novel pathway for the activation of an effective anti-GBM immune response mediated by high-mobility-group box 1 (HMGB1), an alarmin protein released from dying tumor cells, which acts as an endogenous ligand for Toll-like receptor 2 (TLR2) signaling on bone marrow-derived GBM-infiltrating DCs. METHODS AND FINDINGS: Using a combined immunotherapy/conditional cytotoxic approach that utilizes adenoviral vectors (Ad) expressing Fms-like tyrosine kinase 3 ligand (Flt3L) and thymidine kinase (TK) delivered into the tumor mass, we demonstrated that CD4(+) and CD8(+) T cells were required for tumor regression and immunological memory. Increased numbers of bone marrow-derived, tumor-infiltrating myeloid DCs (mDCs) were observed in response to the therapy. Infiltration of mDCs into the GBM, clonal expansion of antitumor T cells, and induction of an effective anti-GBM immune response were TLR2 dependent. We then proceeded to identify the endogenous ligand responsible for TLR2 signaling on tumor-infiltrating mDCs. We demonstrated that HMGB1 was released from dying tumor cells, in response to Ad-TK (+ gancyclovir [GCV]) treatment. Increased levels of HMGB1 were also detected in the serum of tumor-bearing Ad-Flt3L/Ad-TK (+GCV)-treated mice. Specific activation of TLR2 signaling was induced by supernatants from Ad-TK (+GCV)-treated GBM cells; this activation was blocked by glycyrrhizin (a specific HMGB1 inhibitor) or with antibodies to HMGB1. HMGB1 was also released from melanoma, small cell lung carcinoma, and glioma cells treated with radiation or temozolomide. Administration of either glycyrrhizin or anti-HMGB1 immunoglobulins to tumor-bearing Ad-Flt3L and Ad-TK treated mice, abolished therapeutic efficacy, highlighting the critical role played by HMGB1-mediated TLR2 signaling to elicit tumor regression. Therapeutic efficacy of Ad-Flt3L and Ad-TK (+GCV) treatment was demonstrated in a second glioma model and in an intracranial melanoma model with concomitant increases in the levels of circulating HMGB1. CONCLUSIONS: Our data provide evidence for the molecular and cellular mechanisms that support the rationale for the clinical implementation of antibrain cancer immunotherapies in combination with tumor killing approaches in order to elicit effective antitumor immune responses, and thus, will impact clinical neuro-oncology practice.


Subject(s)
Brain Neoplasms/metabolism , HMGB1 Protein/metabolism , Toll-Like Receptor 2/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cell Line, Tumor , Cells, Cultured , Female , Flow Cytometry , Genetic Vectors , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Mice , Mice, Transgenic
15.
Genesis ; 45(4): 184-93, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17417787

ABSTRACT

Many developing tissues require programmed cell death (PCD) for proper formation. In mice and C. elegans, developmental PCD is regulated by the Bcl-2 family of proteins. Two bcl-2 genes are encoded in the Drosophila genome (debcl/dBorg1/Drob-1/dBok and buffy/dBorg2) and previous RNAi-based studies suggested a requirement for these in embryonic development. However, we report here that, despite the fact that many tissues in fruit flies are shaped by PCD, deletion of the bcl-2 genes does not perturb normal development. We investigated whether the fly bcl-2 genes regulate non-apoptotic processes that require caspases, but found these to be bcl-2 gene-independent. However, irradiation of the mutants demonstrates that DNA damage-induced apoptosis, mediated by Reaper, is blocked by buffy and that debcl is required to inhibit buffy. Our results demonstrate that developmental PCD regulation in the fly does not rely upon the Bcl-2 proteins, but that they provide an added layer of protection in the apoptotic response to stress.


Subject(s)
Apoptosis , DNA Damage/physiology , Drosophila/embryology , Proto-Oncogene Proteins c-bcl-2/physiology , Alleles , Animals , Apoptosis/radiation effects , Cell Count , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Embryo, Nonmammalian , Female , Fertility , Male , Membrane Proteins/genetics , Mitosis/genetics , Mutant Proteins/genetics , Mutant Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Radiation, Ionizing
SELECTION OF CITATIONS
SEARCH DETAIL
...