Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Mol Imaging Biol ; 22(2): 256-264, 2020 04.
Article in English | MEDLINE | ID: mdl-31240531

ABSTRACT

In light of the United States Food and Drug Administration (FDA) requirement of 21 CFR 212 current Good Manufacturing Practice (cGMP) for FDA-approved position emission tomography (PET) drugs, the University of California Los Angeles (UCLA) Biomedical Cyclotron (BMC) transformed from a pre-cGMP era academic cyclotron and radiochemistry facility to a current cGMP-compliant PET drug manufacturer. In this article, we share the financial and regulatory compliance aspects of the "transformation" required to develop a sustainable quality system to support the production of two PET drugs under Abbreviated New Drug Applications (ANDAs).


Subject(s)
Drug Industry/standards , Facility Regulation and Control/standards , Guideline Adherence , Positron-Emission Tomography/standards , Radiochemistry/methods , California , Cyclotrons , Drug Approval , Humans , Quality Control , Radiopharmaceuticals , United States , United States Food and Drug Administration , Universities
2.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23403069

ABSTRACT

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Subject(s)
Brain Neoplasms/drug therapy , Clinical Trials, Phase I as Topic/methods , Cytotoxins/administration & dosage , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioblastoma/drug therapy , Immunization/methods , Adenoviridae/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cytotoxins/adverse effects , Cytotoxins/metabolism , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Genetic Vectors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Rats , Rats, Inbred Lew , Tissue Distribution/drug effects , Tissue Distribution/physiology , Treatment Outcome
3.
Hum Gene Ther Methods ; 23(4): 271-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22950971

ABSTRACT

Adenoviral vectors (Ads) have been evaluated in clinical trials for glioma. However, systemic immunity against the vectors can hamper therapeutic efficacy. We demonstrated that combined immunostimulation and cytotoxic gene therapy provides long-term survival in preclinical glioma models. Because helper-dependent high-capacity Ads (HC-Ads) elicit sustained transgene expression, in the presence of antiadenoviral immunity, we engineered HC-Ads encoding conditional cytotoxic herpes simplex type 1 thymidine kinase and immunostimulatory cytokine Fms-like tyrosine kinase ligand-3 under the control of the TetOn system. Escalating doses of combined HC-Ads (1×10(8), 1×10(9), and 1×10(10) viral particles [VP]) were delivered into the rat brain. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points after vector delivery. Histopathological analysis did not reveal any evidence of toxicity or long-term inflammation at the lower doses tested. Vector genomes were restricted to the injection site. Serum chemistry did not uncover adverse systemic side effects at any of the doses tested. Taken together, our data indicate that doses of up to 1×10(9) VP of each HC-Ad can be safely administered into the normal brain. This comprehensive toxicity and biodistribution study will lay the foundations for implementation of a phase 1 clinical trial for GBM using HC-Ads.


Subject(s)
Adenoviridae/genetics , Brain/metabolism , Genetic Vectors/metabolism , Animals , Behavior, Animal/drug effects , Blood Chemical Analysis , Brain/drug effects , Brain/pathology , Clinical Trials, Phase I as Topic , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/toxicity , Glioma/therapy , Herpesvirus 1, Human/enzymology , Humans , Male , Rats , Rats, Inbred Lew , Thymidine Kinase/genetics , Tissue Distribution , Transduction, Genetic , fms-Like Tyrosine Kinase 3/genetics
4.
Neoplasia ; 14(8): 757-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22952428

ABSTRACT

Adenovirus-mediated delivery of the immune-stimulatory cytokine Flt3L and the conditionally cytotoxic thymidine kinase (TK) induces tumor regression and long-term survival in preclinical glioma (glioblastoma multiforme [GBM]) models. Flt3L induces expansion and recruitment of plasmacytoid dendritic cells (pDCs) into the brain. Although pDCs can present antigen and produce powerful inflammatory cytokines, that is, interferon α (IFN-α), their role in tumor immunology remains debated. Thus, we studied the role of pDCs and IFN-α in Ad.TK/GCV+ Ad.Flt3L-mediated anti-GBM therapeutic efficacy. Our data indicate that the combined gene therapy induced recruitment of plasmacytoid DCs (pDCs) into the tumor mass; which were capable of in vivo phagocytosis, IFN-α release, and T-cell priming. Thus, we next used either pDCs or an Ad vector encoding IFN-α delivered within the tumor microenvironment. When rats were treated with Ad.TK/GCV in combination with pDCs or Ad-IFN-α, they exhibited 35% and 50% survival, respectively. However, whereas intracranial administration of Ad.TK/GCV + Ad.Flt3L exhibited a high safety profile, Ad-IFN-α led to severe local inflammation, with neurologic and systemic adverse effects. To elucidate whether the efficacy of the immunotherapy was dependent on IFN-α-secreting pDCs, we administered an Ad vector encoding B18R, an IFN-α antagonist, which abrogated the antitumoral effect of Ad.TK/GCV + Ad.Flt3L. Our data suggest that IFN-α release by activated pDCs plays a critical role in the antitumor effect mediated by Ad.TK/GCV + Ad.Flt3L. In summary, taken together, our results demonstrate that pDCs mediate anti-GBM therapeutic efficacy through the production of IFN-α, thus manipulation of pDCs constitutes an attractive new therapeutic target for the treatment of GBM.


Subject(s)
Brain Neoplasms/immunology , Dendritic Cells/immunology , Glioblastoma/immunology , Interferon-alpha/immunology , Tumor Microenvironment , Adenoviridae/genetics , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/pathology , Glioblastoma/therapy , Immunotherapy , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/genetics , Rats , T-Lymphocytes/immunology , Thymidine Kinase/genetics
5.
Neurotherapeutics ; 9(4): 827-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996231

ABSTRACT

Immune-mediated gene therapy using adenovirus expressing Flt3 ligand and thymidine kinase followed by ganciclovir administration (Flt3/TK) effectively elicits tumor regression in preclinical glioma models. Herein, we assessed new strategies to optimize Flt3L/TK therapeutic efficacy in a refractory RG2 orthotopic glioblastoma model. Specifically, we aimed to optimize the therapeutic efficacy of Flt3L/TK treatment in the RG2 model by overexpressing the following genes within the brain tumor microenvironment: 1) a TK mutant with enhanced cytotoxicity (SR39 mutant TK), 2) Flt3L-IgG fusion protein that has a longer half-life, 3) CD40L to stimulate DC maturation, 4) T helper cell type 1 polarizing dendritic cell cytokines interleukin-12 or C-X-C motif ligand 10 chemokine (CXCL)-10, 5) C-C motif ligand 2 chemokine (CCL2) or C-C motif ligand 3 chemokine (CCL3) to enhance dendritic cell recruitment into the tumor microenvironment, 6) T helper cell type 1 cytokines interferon-γ or interleukin-2 to enhance effector T-cell functions, and 7) IκBα or p65RHD (nuclear factor kappa-B [NF-κB] inhibitors) to suppress the function of Foxp3+ Tregs and enhanced effector T-cell functions. Anti-tumor immunity and tumor specific effector T-cell functions were assessed by cytotoxic T lymphocyte assay and intracellular IFN-γ staining. Our data showed that overexpression of interferon-γ or interleukin-2, or inhibition of the nuclear factor kappa-B within the tumor microenvironment, enhanced cytotoxic T lymphocyte-mediated immune responses and successfully extended the median survival of rats bearing intracranial RG2 when combined with Flt3L/TK. These findings indicate that enhancement of T-cell functions constitutes a critical therapeutic target to overcome immune evasion and enhance therapeutic efficacy for brain cancer. In addition, our study provides novel targets to be used in combination with immune-therapeutic strategies for glioblastoma, which are currently being tested in the clinic.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/therapy , Immunotherapy/methods , Signal Transduction , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Antiviral Agents/therapeutic use , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Dendritic Cells/immunology , Disease Models, Animal , Ganciclovir/therapeutic use , Genetic Vectors , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Interleukin-2/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/therapeutic use , NF-kappa B/immunology , Rats , Recombinant Proteins/therapeutic use , Thymidine Kinase/therapeutic use , Tumor Microenvironment/immunology
6.
Proc Natl Acad Sci U S A ; 109(20): 7835-40, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22547816

ABSTRACT

Following antigen recognition on target cells, effector T cells establish immunological synapses and secrete cytokines. It is thought that T cells secrete cytokines in one of two modes: either synaptically (i.e., toward antigenic target cells) or multidirectionally, affecting a wider population of cells. This paradigm predicts that synaptically secreted cytokines such as IFN-γ will preferentially signal to antigenic target cells contacted by the T cell through an immunological synapse. Despite its physiological significance, this prediction has never been tested. We developed a live-cell imaging system to compare the responses of target cells and nonantigenic bystanders to IFN-γ secreted by CD8+, antigen-specific, cytotoxic T cells. Both target cells and surrounding nontarget cells respond robustly. This pattern of response was detected even at minimal antigenic T-cell stimulation using low doses of antigenic peptide, or altered peptide ligands. Although cytotoxic immunological synapses restrict killing to antigenic target cells, the effects of IFN-γ are more widespread.


Subject(s)
Immunological Synapses/immunology , Interferon-gamma/metabolism , T-Lymphocytes, Cytotoxic/immunology , Adenoviridae , Analysis of Variance , Astrocytes/immunology , Genetic Vectors/genetics , Green Fluorescent Proteins , Image Processing, Computer-Assisted , Interferon-gamma/immunology , Microscopy/methods
7.
Mol Ther ; 20(4): 808-19, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22233583

ABSTRACT

The adaptive immune response to viral vectors reduces vector-mediated transgene expression from the brain. It is unknown, however, whether this loss is caused by functional downregulation of transgene expression or death of transduced cells. Herein, we demonstrate that during the elimination of transgene expression, the brain becomes infiltrated with CD4(+) and CD8(+) T cells and that these T cells are necessary for transgene elimination. Further, the loss of transgene-expressing brain cells fails to occur in the absence of IFNγ, perforin, and TNFα receptor. Two methods to induce severe immune suppression in immunized animals also fail to restitute transgene expression, demonstrating the irreversibility of this process. The need for cytotoxic molecules and the irreversibility of the reduction in transgene expression suggested to us that elimination of transduced cells is responsible for the loss of transgene expression. A new experimental paradigm that discriminates between downregulation of transgene expression and the elimination of transduced cells demonstrates that transduced cells are lost from the brain upon the induction of a specific antiviral immune response. We conclude that the anti-adenoviral immune response reduces transgene expression in the brain through loss of transduced cells.


Subject(s)
Brain/cytology , Interferon-gamma/metabolism , Perforin/metabolism , Transduction, Genetic/methods , Transgenes/genetics , Tumor Necrosis Factor-alpha/metabolism , Adenoviridae/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic
8.
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
9.
PLoS One ; 6(8): e23523, 2011.
Article in English | MEDLINE | ID: mdl-21897844

ABSTRACT

CD8(+) T cells infiltrate the brain during an anti-viral immune response. Within the brain CD8(+) T cells recognize cells expressing target antigens, become activated, and secrete IFNγ. However, there are no methods to recognize individual cells that respond to IFNγ. Using a model that studies the effects of the systemic anti-adenoviral immune response upon brain cells infected with an adenoviral vector in mice, we describe a method that identifies individual cells that respond to IFNγ. To identify individual mouse brain cells that respond to IFNγ we constructed a series of adenoviral vectors that contain a transcriptional response element that is selectively activated by IFNγ signaling, the gamma-activated site (GAS) promoter element; the GAS element drives expression of a transgene, Cre recombinase (Ad-GAS-Cre). Upon binding of IFNγ to its receptor, the intracellular signaling cascade activates the GAS promoter, which drives expression of the transgene Cre recombinase. We demonstrate that upon activation of a systemic immune response against adenovirus, CD8(+) T cells infiltrate the brain, interact with target cells, and cause an increase in the number of cells expressing Cre recombinase. This method can be used to identify, study, and eventually determine the long term fate of infected brain cells that are specifically targeted by IFNγ. The significance of this method is that it will allow to characterize the networks in the brain that respond to the specific secretion of IFNγ by anti-viral CD8(+) T cells that infiltrate the brain. This will allow novel insights into the cellular and molecular responses underlying brain immune responses.


Subject(s)
Antiviral Agents/metabolism , Brain/cytology , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/metabolism , Molecular Imaging/methods , Signal Transduction/immunology , Adenoviridae/genetics , Animals , Brain/metabolism , Brain/virology , Female , Genes, Reporter/genetics , Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Integrases/metabolism , Interferon-gamma/genetics , Male , Mice , Promoter Regions, Genetic/genetics , Species Specificity , Thymidine Kinase/genetics
10.
Clin Cancer Res ; 17(14): 4705-18, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21632862

ABSTRACT

PURPOSE: Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Clinical trials for GBM using dendritic cell (DC) vaccination resulted in antitumor immune responses. Herein, we tested the hypothesis that combining in situ (intratumoral) Ad-Flt3L/Ad-TK-mediated gene therapy with DC vaccination would increase therapeutic efficacy and antitumor immunity. EXPERIMENTAL DESIGN: We first assessed the immunogenicity of tumor lysates generated by Ad-TK (+GCV), temozolomide (TMZ), or freeze/thawing cycles (FTC) in a syngeneic brain tumor model. We also assessed phenotypic markers, cytokine release, and phagocytosis of bone marrow-derived DCs generated by fms-like tyrosine kinase 3 ligand (Flt3L) + IL-6 or by granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL) 4. Inhibition of tumor progression and production of anti-GBM antibodies was assessed following vaccination with (i) tumor cell lysates, (ii) DCs generated with either Flt3L/IL-6 or GM-CSF/IL-4 loaded with either Ad-TK/GCV-, TMZ-, or FTC-generated tumor lysates, or (iii) DCs in combination with in situ Ad-Flt3L/Ad-TK gene therapy. RESULTS: DCs loaded with tumor cell lysates generated with either Ad-TK/GCV or TMZ led to increased levels of phagocytosis, therapeutic efficacy, and humoral immune response. In situ immunogene therapy in combination with DC vaccination led to brain tumor regression and long-term survival in about 90% of animals, a significant increase when compared with either therapy alone. CONCLUSIONS: Our results indicate that modifying the tumor microenvironment using intratumoral Ad-Flt3L/Ad-TK-mediated gene therapy potentiates therapeutic efficacy and antitumor immunity induced by DC vaccination. These data support novel phase I clinical trials to assess the safety and efficacy of this combined approach.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Glioblastoma/therapy , Tumor Microenvironment/immunology , Adenoviridae/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Cell Death/immunology , Cell Line, Tumor , Clinical Trials as Topic , Cytokines/metabolism , Dendritic Cells/transplantation , Disease Models, Animal , Gene Expression Regulation, Viral , Genetic Therapy , Genetic Vectors/genetics , Glioblastoma/genetics , Glioblastoma/immunology , Rats , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tumor Microenvironment/genetics
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.
Mol Ther ; 19(10): 1793-801, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21505426

ABSTRACT

Glioblastoma multiforme (GBM) is a primary brain tumor with a median survival of 14.6 months postdiagnosis. The infiltrative nature of GBM prevents complete resection and residual brain tumor cells give rise to recurrent GBM, a hallmark of this disease. Recurrent GBMs are known to harbor numerous mutations/gene rearrangements when compared to the primary tumor, which leads to the potential expression of novel proteins that could serve as tumor neoantigens. We have developed a combined immune-based gene therapeutic approach for GBM using adenoviral (Ads) mediated gene delivery of Herpes Simplex Virus Type 1-thymidine kinase (TK) into the tumor mass to induce tumor cells' death combined with an adenovirus expressing fms-like tyrosine kinase 3 ligand (Flt3L) to recruit dendritic cells (DCs) into the tumor microenvironment. This leads to the induction of specific anti-brain tumor immunity and immunological memory. In a model of GBM recurrence, we demonstrate that Flt3L/TK mediated immunological memory is capable of recognizing brain tumor neoantigens absent from the original treated tumor. These data demonstrate that the Flt3L/TK gene therapeutic approach can induce systemic immunological memory capable of recognizing a brain tumor neoantigen in a model of recurrent GBM.


Subject(s)
Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Thymidine Kinase/genetics , fms-Like Tyrosine Kinase 3/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunologic Memory , Interferon-gamma/metabolism , T-Lymphocytes/immunology
14.
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
15.
Proc Natl Acad Sci U S A ; 107(46): 20021-6, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21030678

ABSTRACT

Restricting the cytotoxicity of anticancer agents by targeting receptors exclusively expressed on tumor cells is critical when treating infiltrative brain tumors such as glioblastoma multiforme (GBM). GBMs express an IL-13 receptor (IL13Rα2) that differs from the physiological IL4R/IL13R receptor. We developed a regulatable adenoviral vector (Ad.mhIL-4.TRE.mhIL-13-PE) encoding a mutated human IL-13 fused to Pseudomonas exotoxin (mhIL-13-PE) that specifically binds to IL13Rα2 to provide sustained expression, effective anti-GBM cytotoxicity, and minimal neurotoxicity. The therapeutic Ad also encodes mutated human IL-4 that binds to the physiological IL4R/IL13R without interacting with IL13Rα2, thus inhibiting potential binding of mhIL-13-PE to normal brain cells. Using intracranial GBM xenografts and syngeneic mouse models, we tested the Ad.mhIL-4.TRE.mhIL-13-PE and two protein formulations, hIL-13-PE used in clinical trials (Cintredekin Besudotox) and a second-generation mhIL-13-PE. Cintredekin Besudotox doubled median survival without eliciting long-term survival and caused severe neurotoxicity; mhIL-13-PE led to ∼40% long-term survival, eliciting severe neurological toxicity at the high dose tested. In contrast, Ad-mediated delivery of mhIL-13-PE led to tumor regression and long-term survival in over 70% of the animals, without causing apparent neurotoxicity. Although Cintredekin Besudotox was originally developed to target GBM, when tested in a phase III trial it failed to achieve clinical endpoints and revealed neurotoxicity. Limitations of Cintredekin Besudotox include its short half-life, which demanded frequent or continued administration, and binding to IL4R/IL13R, present in normal brain cells. These shortcomings were overcome by our therapeutic Ad, thus representing a significant advance in the development of targeted therapeutics for GBM.


Subject(s)
Brain Neoplasms/drug therapy , Cytotoxins/genetics , Cytotoxins/therapeutic use , Gene Transfer Techniques , Genetic Therapy , Glioma/drug therapy , Adenoviridae/genetics , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Exotoxins/genetics , Exotoxins/therapeutic use , Genetic Vectors/genetics , Glioma/pathology , Humans , Immunocompetence/immunology , Interleukin-13/genetics , Interleukin-13/therapeutic use , Mice , Mice, Nude , Mutation/genetics , Neurotoxins/toxicity , Pseudomonas/metabolism , Transgenes/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
16.
Proc Natl Acad Sci U S A ; 107(32): 14443-8, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660723

ABSTRACT

Soluble antigens diffuse out of the brain and can thus stimulate a systemic immune response, whereas particulate antigens (from infectious agents or tumor cells) remain within brain tissue, thus failing to stimulate a systemic immune response. Immune privilege describes how the immune system responds to particulate antigens localized selectively within the brain parenchyma. We believe this immune privilege is caused by the absence of antigen presenting dendritic cells from the brain. We tested the prediction that expression of fms-like tyrosine kinase ligand 3 (Flt3L) in the brain will recruit dendritic cells and induce a systemic immune response against exogenous influenza hemagglutinin in BALB/c mice. Coexpression of Flt3L with HA in the brain parenchyma induced a robust systemic anti-HA immune response, and a small response against myelin basic protein and proteolipid protein epitopes. Depletion of CD4(+)CD25+ regulatory T cells (Tregs) enhanced both responses. To investigate the autoimmune impact of these immune responses, we characterized the neuropathological and behavioral consequences of intraparenchymal injections of Flt3L and HA in BALB/c and C57BL/6 mice. T cell infiltration in the forebrain was time and strain dependent, and increased in animals treated with Flt3L and depleted of Tregs; however, we failed to detect widespread defects in myelination throughout the forebrain or spinal cord. Results of behavioral tests were all normal. These results demonstrate that Flt3L overcomes the brain's immune privilege, and supports the clinical development of Flt3L as an adjuvant to stimulate clinically effective immune responses against brain neo-antigens, for example, those associated with brain tumors.


Subject(s)
Brain/immunology , Immune System/immunology , fms-Like Tyrosine Kinase 3/immunology , Adjuvants, Immunologic , Animals , Antigens/immunology , Dendritic Cells/immunology , Hemagglutinins/immunology , Immunity , Ligands , Mice , Mice, Inbred BALB C , Prosencephalon/immunology , Spinal Cord/immunology , T-Lymphocytes, Regulatory/immunology
17.
J Virol ; 84(12): 6007-17, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20375153

ABSTRACT

Glioblastoma multiforme (GBM) is a deadly primary brain tumor. Conditional cytotoxic/immune-stimulatory gene therapy (Ad-TK and Ad-Flt3L) elicits tumor regression and immunological memory in rodent GBM models. Since the majority of patients enrolled in clinical trials would exhibit adenovirus immunity, which could curtail transgene expression and therapeutic efficacy, we used high-capacity adenovirus vectors (HC-Ads) as a gene delivery platform. Herein, we describe for the first time a novel bicistronic HC-Ad driving constitutive expression of herpes simplex virus type 1 thymidine kinase (HSV1-TK) and inducible Tet-mediated expression of Flt3L within a single-vector platform. We achieved anti-GBM therapeutic efficacy with no overt toxicities using this bicistronic HC-Ad even in the presence of systemic Ad immunity. The bicistronic HC-Ad-TK/TetOn-Flt3L was delivered into intracranial gliomas in rats. Survival, vector biodistribution, neuropathology, systemic toxicity, and neurobehavioral deficits were assessed for up to 1 year posttreatment. Therapeutic efficacy was also assessed in animals preimmunized against Ads. We demonstrate therapeutic efficacy, with vector genomes being restricted to the brain injection site and an absence of overt toxicities. Importantly, antiadenoviral immunity did not inhibit therapeutic efficacy. These data represent the first report of a bicistronic vector platform driving the expression of two therapeutic transgenes, i.e., constitutive HSV1-TK and inducible Flt3L genes. Further, our data demonstrate no promoter interference and optimum gene delivery and expression from within this single-vector platform. Analysis of the efficacy, safety, and toxicity of this bicistronic HC-Ad vector in an animal model of GBM strongly supports further preclinical testing and downstream process development of HC-Ad-TK/TetOn-Flt3L for a future phase I clinical trial for GBM.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Glioma/genetics , Glioma/therapy , Herpesvirus 1, Human/enzymology , Thymidine Kinase/therapeutic use , Viral Proteins/therapeutic use , fms-Like Tyrosine Kinase 3/therapeutic use , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glioma/metabolism , Herpesvirus 1, Human/genetics , Humans , Rats , Rats, Inbred Lew , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
18.
Curr Protoc Neurosci ; Chapter 4: Unit 4.24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20066657

ABSTRACT

Viral vector-mediated gene delivery is an attractive procedure for introducing genes into the brain, both for purposes of basic neuroscience research and to develop gene therapy for neurological diseases. Replication-defective adenoviruses possess many features which make them ideal vectors for this purpose-efficiently transducing terminally differentiated cells such as neurons and glial cells, resulting in high levels of transgene expression in vivo. Also, in the absence of anti-adenovirus immunity, these vectors can sustain very long-term transgene expression within the brain parenchyma. This unit provides protocols for the stereotactic injection of adenoviral vectors into the brain, followed by protocols to detect transgene expression or infiltrates of immune cells by immunocytochemistry or immunofluorescence. ELISPOT and neutralizing antibody assay methodologies are provided to quantitate the levels of cellular and humoral immune responses against adenoviruses. Quantitation of adenoviral vector genomes within the rat brain using qPCR is also described.


Subject(s)
Adenoviridae/genetics , Brain/virology , Gene Transfer Techniques , Genetic Vectors , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Fluorescent Antibody Technique/methods , Gene Transfer Techniques/adverse effects , Genetic Vectors/adverse effects , Immunohistochemistry/methods , Neuroimmunomodulation , Polymerase Chain Reaction/methods , Rats , Stereotaxic Techniques
19.
Clin Cancer Res ; 15(19): 6113-27, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789315

ABSTRACT

PURPOSE: Glioblastoma multiforme is a deadly primary brain cancer. Because the tumor kills due to recurrences, we tested the hypothesis that a new treatment would lead to immunological memory in a rat model of recurrent glioblastoma multiforme. EXPERIMENTAL DESIGN: We developed a combined treatment using an adenovirus (Ad) expressing fms-like tyrosine kinase-3 ligand (Flt3L), which induces the infiltration of immune cells into the tumor microenvironment, and an Ad expressing herpes simplex virus-1-thymidine kinase (TK), which kills proliferating tumor cells in the presence of ganciclovir. RESULTS: This treatment induced immunological memory that led to rejection of a second glioblastoma multiforme implanted in the contralateral hemisphere and of an extracranial glioblastoma multiforme implanted intradermally. Rechallenged long-term survivors exhibited anti-glioblastoma multiforme-specific T cells and displayed specific delayed-type hypersensitivity. Using depleting antibodies, we showed that rejection of the second tumor was dependent on CD8(+) T cells. Circulating anti-glioma antibodies were observed when glioblastoma multiforme cells were implanted intradermally in naïve rats or in long-term survivors. However, rats bearing intracranial glioblastoma multiforme only exhibited circulating antitumoral antibodies upon treatment with Ad-Flt3L + Ad-TK. This combined treatment induced tumor regression and release of the chromatin-binding protein high mobility group box 1 in two further intracranial glioblastoma multiforme models, that is, Fisher rats bearing intracranial 9L and F98 glioblastoma multiforme cells. CONCLUSIONS: Treatment with Ad-Flt3L + Ad-TK triggered systemic anti-glioblastoma multiforme cellular and humoral immune responses, and anti-glioblastoma multiforme immunological memory. Release of the chromatin-binding protein high mobility group box 1 could be used as a noninvasive biomarker of therapeutic efficacy for glioblastoma multiforme. The robust treatment efficacy lends further support to its implementation in a phase I clinical trial.


Subject(s)
Brain Neoplasms/therapy , Cytotoxicity, Immunologic/genetics , Genetic Therapy/methods , Glioma/therapy , Immunity, Cellular/genetics , Immunity, Humoral/genetics , Immunologic Memory/genetics , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Genes, Transgenic, Suicide/genetics , Genes, Transgenic, Suicide/physiology , Glioma/immunology , Glioma/pathology , Humans , Immunotherapy/methods , Lymphocyte Activation/genetics , Membrane Proteins/genetics , Models, Biological , Organ Specificity/genetics , Organ Specificity/immunology , Rats , Remission Induction/methods , Thymidine Kinase/genetics , Tumor Burden
20.
Curr Gene Ther ; 9(5): 409-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19860655

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain cancer in adults. Despite significant advances in treatment and intensive research, the prognosis for patients with GBM remains poor. Therapeutic challenges for GBM include its invasive nature, the proximity of the tumor to vital brain structures often preventing total resection, and the resistance of recurrent GBM to conventional radiotherapy and chemotherapy. Gene therapy has been proposed as a useful adjuvant for GBM, to be used in conjunction with current treatment. Work from our laboratory has shown that combination of conditional cytotoxic with immunotherapeutic approaches for the treatment of GBM elicits regression of large intracranial tumor masses and anti-tumor immunological memory in syngeneic rodent models of GBM. In this review we examined the currently available animal models for GBM, including rodent transplantable models, endogenous rodent tumor models and spontaneous GBM in dogs. We discuss non-invasive surrogate end points to assess tumor progression and therapeutic efficacy, such as behavioral tests and circulating biomarkers. Growing preclinical and clinical data contradict the old dogma that cytotoxic anti-cancer therapy would lead to an immune-suppression that would impair the ability of the immune system to mount an anti-tumor response. The implications of the findings reviewed indicate that combination of cytotoxic therapy with immunotherapy will lead to synergistic antitumor efficacy with reduced neurotoxicity and supports the clinical implementation of combined cytotoxic-immunotherapeutic strategies for the treatment of patients with GBM.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Animals , Behavior, Animal , Combined Modality Therapy , Disease Models, Animal , Dogs , Humans , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...