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 ; 12(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38724464

ABSTRACT

BACKGROUND: Glioblastoma (GBM) almost invariably becomes resistant towards conventional treatment of radiotherapy and temozolomide (TMZ) chemotherapy, partly due to subpopulations of intrinsically resistant glioma stem-like cells (GSC). The oncolytic herpes simplex virus-1 G207 is a promising approach for GBM virotherapy although its efficacy in patients with GBM is often limited. Natural killer group 2 member D ligands (NKG2DLs) are minimally expressed by healthy cells but are upregulated by the DNA damage response (DDR) and in malignant cells with chronic DDR signaling, resulting in innate immune activation. METHODS: We have designed a bispecific T-cell engager (BiTE) capable of cross-linking CD3 on T cells with NKG2DL-expressing GBM cells. We then engineered the G207 virus to express the NKG2D BiTE and secrete it from infected cells. The efficacy of the free BiTE and BiTE delivered by G207 was evaluated in combination with conventional therapies in GBM cells and against patient-derived GSCs in the context of T-cell activation and target cell viability. RESULTS: NKG2D BiTE-mediated cross-linking of GBM cells and T cells causes antigen-independent T-cell activation, pro-inflammatory cytokine release, and tumor cell death, thereby combining direct viral oncolysis with BiTE-mediated cytotoxicity. Surface NKG2DL expression was further elevated on GBM cells following pretreatment with sublethal doses of TMZ and radiation to induce the DDR, increasing sensitivity towards G207-NKG2D BiTE and achieving synergistic cytotoxicity. We also demonstrate a novel strategy for targeting GSCs that are non-permissive to G207 infection but remain sensitive to NKG2D BiTE. CONCLUSIONS: We propose a potential model for targeting GSCs in heterogeneous tumors, whereby differentiated GBM cells infected with G207-NKG2D BiTE produce NKG2D BiTE locally, directing T-cell cytotoxicity towards the GSC subpopulations in the tumor microenvironment.


Subject(s)
Glioblastoma , NK Cell Lectin-Like Receptor Subfamily K , Neoplastic Stem Cells , Oncolytic Virotherapy , Humans , Glioblastoma/therapy , Glioblastoma/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplastic Stem Cells/metabolism , Oncolytic Virotherapy/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor
2.
J Immunother Cancer ; 9(4)2021 03.
Article in English | MEDLINE | ID: mdl-33820820

ABSTRACT

BACKGROUND: Programmed death-ligand 1 (PD-L1) is an important immune checkpoint protein that can be regarded as a pan-cancer antigen expressed by multiple different cell types within the tumor. While antagonizing PD-L1 is well known to relieve PD-1/PD-L1-mediated T cell suppression, here we have combined this approach with an immunotherapy strategy to target T cell cytotoxicity directly toward PD-L1-expressing cells. We developed a bi-specific T cell engager (BiTE) crosslinking PD-L1 and CD3ε and demonstrated targeted cytotoxicity using a clinically relevant patient-derived ascites model. This approach represents an immunological 'volte-face' whereby a tumor immunological defense mechanism can be instantly transformed into an Achilles' heel for targeted immunotherapy. METHODS: The PD-L1 targeting BiTE comprises an anti-PD-L1 single-chain variable fragment (scFv) or nanobody (NB) domain and an anti-CD3 scFv domain in a tandem repeat. The ability to activate T cell cytotoxicity toward PD-L1-expressing cells was established using human carcinoma cells and PD-L1-expressing human ('M2') macrophages in the presence of autologous T cells. Furthermore, we armed oncolytic herpes simplex virus-1 (oHSV-1) with PD-L1 BiTE and demonstrated successful delivery and targeted cytotoxicity in unpurified cultures of malignant ascites derived from different cancer patients. RESULTS: PD-L1 BiTE crosslinks PD-L1-positive cells and CD3ε on T cells in a 'pseudo-synapse' and triggers T cell activation and release of proinflammatory cytokines such as interferon-gamma (IFN-γ), interferon gamma-induced protein 10 (IP-10) and tumour necrosis factor-α (TNF-α). Activation of endogenous T cells within ascites samples led to significant lysis of tumor cells and M2-like macrophages (CD11b+CD64+ and CD206+/CD163+). The survival of CD3+ T cells (which can also express PD-L1) was unaffected. Intriguingly, ascites fluid that appeared particularly immunosuppressive led to higher expression of PD-L1 on tumor cells, resulting in improved BiTE-mediated T cell activation. CONCLUSIONS: The study reveals that PD-L1 BiTE is an effective immunotherapeutic approach to kill PD-L1-positive tumor cells and macrophages while leaving T cells unharmed. This approach activates endogenous T cells within malignant ascites, generates a proinflammatory response and eliminates cells promoting tumor progression. Using an oncolytic virus for local expression of PD-L1 BiTE also prevents 'on-target off-tumor' systemic toxicities and harnesses immunosuppressive protumor conditions to augment immunotherapy in immunologically 'cold' clinical cancers.


Subject(s)
Antibodies, Bispecific/immunology , B7-H1 Antigen/immunology , CD3 Complex/immunology , Herpesvirus 1, Human/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , B7-H1 Antigen/metabolism , CD3 Complex/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Coculture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , HEK293 Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/virology , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , T-Lymphocytes/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Vero Cells
3.
J Virol Methods ; 291: 114099, 2021 05.
Article in English | MEDLINE | ID: mdl-33592218

ABSTRACT

The development of a T7 RNA polymerase (T7 RNAP) expressing cell line i.e. BSR T7/5 cells marks an improvement of reverse genetics for the recovery of recombinant Newcastle disease virus (rNDV). BSR T7/5 is developed by transient transfection of plasmid encoding T7 RNAP gene for rNDV rescue. However, the gene expression decreases gradually over multiple passages and eventually hinders the rescue of rNDV. To address this issue, lentiviral vector was used to develop T7 RNAP-expressing HEK293-TA (HEK293-TA-Lv-T7) and SW620 (SW620-Lv-T7) cell lines, evidenced by the expression of T7 RNAP after subsequent 20 passages. rNDV was rescued successfully using HEK293-TA-Lv-T7 clones (R1D3, R1D8, R5B9) and SW620-Lv-T7 clones (R1C11, R3C5) by reverse transfection, yielding comparable virus rescue efficiency and virus titres to that of BSR T7/5. This study provides new tools for rNDV rescue and insights into cell line development and virology by reverse genetics.


Subject(s)
Lentivirus , Newcastle disease virus , Animals , DNA-Directed RNA Polymerases/genetics , Genetic Vectors , HEK293 Cells , Humans , Lentivirus/genetics , Newcastle disease virus/genetics , Plasmids , Transfection , Viral Proteins
4.
Cancers (Basel) ; 12(4)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32224979

ABSTRACT

Ionising radiation causes cell death through the induction of DNA damage, particularly double-stranded DNA (dsDNA) breaks. Evidence suggests that adenoviruses inhibit proteins involved in the DNA damage response (DDR) to prevent recognition of double-stranded viral DNA genomes as cellular dsDNA breaks. We hypothesise that combining adenovirus treatment with radiotherapy has the potential for enhancing tumour-specific cytotoxicity through inhibition of the DDR and augmentation of virus production. We show that EnAd, an Ad3/Ad11p chimeric oncolytic adenovirus currently being trialled in colorectal and other cancers, targets the DDR pathway at a number of junctures. Infection is associated with a decrease in irradiation-induced 53BP1 and Rad51 foci formation, and in total DNA ligase IV levels. We also demonstrate a radiation-associated increase in EnAd production in vitro and in a pilot in vivo experiment. Given the current limitations of in vitro techniques in assessing for synergy between these treatments, we adapted the plaque assay to allow monitoring of viral plaque size and growth and utilised the xCELLigence cell adhesion assay to measure cytotoxicity. Our study provides further evidence on the interaction between adenovirus and radiation in vitro and in vivo and suggests these have at least an additive, and possibly a synergistic, impact on cytotoxicity.

5.
Infect Genet Evol ; 82: 104287, 2020 08.
Article in English | MEDLINE | ID: mdl-32179148

ABSTRACT

During in vitro selection and evolution screens to adapt the tightly cell-associated bovine foamy virus BFV to high titer cell-free transmission, common, cell-type specific and concurrent adaptive changes in Gag and Env, the major players of foamy virus particle assembly and release, were detected. Upon early establishment of cell type-independent pioneering mutations in Env and, subsequently in Gag, a diverse virus pool emerged that was characterized by the occurrence of shared and additional cell type-specific exchanges. At late passages and saturated titers, remarkably homogeneous virus populations characterized by functionally important mutations developed which may be partly due to stochastic evolutionary events that occurred earlier during adaptation. Reverse genetics showed that defined mutations were functionally important for high titer cell-free transmission.


Subject(s)
Gene Products, env/genetics , Gene Products, gag/genetics , Host-Pathogen Interactions/physiology , Spumavirus/pathogenicity , Adaptation, Biological , Animals , Cattle , Cell Line , Cricetinae , Gene Products, env/metabolism , Gene Products, gag/metabolism , HEK293 Cells , Humans , Retroviridae Infections/transmission , Retroviridae Infections/virology , Reverse Genetics , Virus Assembly
6.
Methods Mol Biol ; 2058: 261-270, 2020.
Article in English | MEDLINE | ID: mdl-31486044

ABSTRACT

The translational success of oncolytic virotherapies would benefit from the widespread use of clinically relevant ex vivo models. Malignant ascites, an accumulation of fluid in the peritoneum due to disseminated cancer, recapitulates many features of the tumor microenvironment, making it a valuable model for studying oncolytic virus activity. Here, we describe a method for the separation and storage of cellular and acellular components of malignant ascites, followed by flow cytometric characterization of the cellular fraction. We then outline a simple experiment using whole ascites to assess the activity of a bispecific T cell engager (BiTE)-expressing oncolytic adenovirus.


Subject(s)
Ascites , Ascitic Fluid , Genetic Vectors , Liquid Biopsy , Oncolytic Virotherapy , Oncolytic Viruses , Adenoviridae/genetics , Biomarkers , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Genetic Vectors/genetics , Humans , Immunophenotyping , Liquid Biopsy/methods , Neoplasms/diagnosis , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics
7.
Mol Ther Oncolytics ; 15: 117-130, 2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31890865

ABSTRACT

Oncolytic viruses represent an emerging approach to cancer therapy. However, better understanding of their interaction with the host cancer cell and approaches to enhance their efficacy are needed. Here, we investigate the effect of chemically induced endoplasmic reticulum (ER) stress on the activity of the chimeric group B adenovirus Enadenotucirev, its closely related parental virus Ad11p, and the archetypal group C oncolytic adenovirus Ad5. We show that treatment of colorectal and ovarian cancer cell lines with thapsigargin or ionomycin caused an influx of Ca2+, leading to an upregulation in E1A transcript and protein levels. Increased E1A protein levels, in turn, increased levels of expression of the E2B viral DNA polymerase, genome replication, late viral protein expression, infectious virus particle production, and cell killing during Enadenotucirev and Ad11p, but not Ad5, infection. This effect was not due to the induction of ER stress, but rather the influx of extracellular Ca2+ and consequent increase in protein kinase C activity. These results underscore the importance of Ca2+ homeostasis during adenoviral infection, indicate a signaling pathway between protein kinase C and E1A, and raise the possibility of using Ca2+ flux-modulating agents in the manufacture and potentiation of oncolytic virotherapies.

8.
Cancer Res ; 78(24): 6852-6865, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30449733

ABSTRACT

: Effective immunotherapy of stromal-rich tumors requires simultaneous targeting of cancer cells and immunosuppressive elements of the microenvironment. Here, we modified the oncolytic group B adenovirus enadenotucirev to express a stroma-targeted bispecific T-cell engager (BiTE). This BiTE bound fibroblast activation protein on cancer-associated fibroblasts (CAF) and CD3ε on T cells, leading to potent T-cell activation and fibroblast death. Treatment of fresh clinical biopsies, including malignant ascites and solid prostate cancer tissue, with FAP-BiTE-encoding virus induced activation of tumor-infiltrating PD1+ T cells to kill CAFs. In ascites, this led to depletion of CAF-associated immunosuppressive factors, upregulation of proinflammatory cytokines, and increased gene expression of markers of antigen presentation, T-cell function, and trafficking. M2-like ascites macrophages exhibited a proinflammatory repolarization, indicating spectrum-wide alteration of the tumor microenvironment. With this approach, we have actively killed both cancer cells and tumor fibroblasts, reversing CAF-mediated immunosuppression and yielding a potent single-agent therapeutic that is ready for clinical assessment. SIGNIFICANCE: An engineered oncolytic adenovirus that encodes a bispecific antibody combines direct virolysis with endogenous T-cell activation to attack stromal fibroblasts, providing a multimodal treatment strategy within a single therapeutic agent.


Subject(s)
Adenoviridae/immunology , Neoplasms/immunology , Neoplasms/metabolism , Oncolytic Viruses/immunology , T-Lymphocytes/immunology , Biopsy , CD3 Complex/metabolism , Coculture Techniques , Combined Modality Therapy , Cytokines/metabolism , Fibroblasts/metabolism , HEK293 Cells , Humans , Immunosuppression Therapy , Inflammation , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...