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.
Viruses ; 13(5)2021 05 08.
Article in English | MEDLINE | ID: mdl-34066836

ABSTRACT

We previously developed a refined, tumor-selective adenovirus, Ad5NULL-A20, harboring tropism ablating mutations in each major capsid protein, to ablate all native means of infection. We incorporated a 20-mer peptide (A20) in the fiber knob for selective infection via αvß6 integrin, a marker of aggressive epithelial cancers. Methods: To ascertain the selectivity of Ad5NULL-A20 for αvß6-positive tumor cell lines of pancreatic and breast cancer origin, we performed reporter gene and cell viability assays. Biodistribution of viral vectors in mice harboring xenografts with low, medium, and high αvß6 levels was quantified by qPCR for viral genomes 48 h post intravenous administration. Results: Ad5NULL-A20 vector transduced cells in an αvß6-selective manner, whilst cell killing mediated by oncolytic Ad5NULL-A20 was αvß6-selective. Biodistribution analysis following intravenous administration into mice bearing breast cancer xenografts demonstrated that Ad5NULL-A20 resulted in significantly reduced liver accumulation coupled with increased tumor accumulation compared to Ad5 in all three models, with tumor-to-liver ratios improved as a function of αvß6 expression. Conclusions: Ad5NULL-A20-based virotherapies efficiently target αvß6-integrin-positive tumors following intravenous administration, validating the potential of Ad5NULL-A20 for systemic applications, enabling tumor-selective overexpression of virally encoded therapeutic transgenes.


Subject(s)
Adenoviridae/genetics , Antigens, Neoplasm/genetics , Genetic Therapy , Genetic Vectors/genetics , Integrins/genetics , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Administration, Intravenous , Animals , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Mice , Neoplasms/etiology , Oncolytic Virotherapy/methods , Transduction, Genetic , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 20(5): 775-786, 2021 05.
Article in English | MEDLINE | ID: mdl-33649105

ABSTRACT

The development of antimetastatic drugs is an urgent healthcare priority for patients with cancer, because metastasis is thought to account for around 90% of cancer deaths. Current antimetastatic treatment options are limited and often associated with poor long-term survival and systemic toxicities. Bcl3, a facilitator protein of the NF-κB family, is associated with poor prognosis in a range of tumor types. Bcl3 has been directly implicated in the metastasis of tumor cells, yet is well tolerated when constitutively deleted in murine models, making it a promising therapeutic target. Here, we describe the identification and characterization of the first small-molecule Bcl3 inhibitor, by using a virtual drug design and screening approach against a computational model of the Bcl3-NF-kB1(p50) protein-protein interaction. From selected virtual screening hits, one compound (JS6) showed potent intracellular Bcl3-inhibitory activity. JS6 treatment led to reductions in Bcl3-NF-kB1 binding, tumor colony formation, and cancer cell migration in vitro; and tumor stasis and antimetastatic activity in vivo, while being devoid of overt systemic toxicity. These results represent a successful application of in silico screening in the identification of protein-protein inhibitors for novel intracellular targets, and confirm Bcl3 as a potential antimetastatic target.


Subject(s)
B-Cell Lymphoma 3 Protein/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude , Models, Molecular
3.
J Immunother Cancer ; 7(1): 280, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31665084

ABSTRACT

BACKGROUND: The treatment of endometrial cancer (EC), the most common gynecological cancer, is currently hampered by the toxicity of current cytotoxic agents, meaning novel therapeutic approaches are urgently required. METHODS: A cohort of 161 patients was evaluated for the expression of the receptor for advanced glycation end products (RAGE) in endometrial tissues. The present study also incorporates a variety of in vitro methodologies within multiple cell lines to evaluate RAGE expression and antibody-drug conjugate efficacy, internalisation and intercellular trafficking. Additionally, we undertook in vivo bio-distribution and toxicity evaluation to determine the suitability of our chosen therapeutic approach, together with efficacy studies in a mouse xenograft model of disease. RESULTS: We have identified an association between over-expression of the receptor for advanced glycation end products (RAGE) and EC (H-score = Healthy: 0.46, SD 0.26; Type I EC: 2.67, SD 1.39; Type II EC: 2.20, SD 1.34; ANOVA, p < 0.0001). Furthermore, increased expression was negatively correlated with patient survival (Spearman's Rank Order Correlation: ρ = - 0.3914, p < 0.05). To exploit this association, we developed novel RAGE-targeting antibody drug conjugates (ADC) and demonstrated the efficacy of this approach. RAGE-targeting ADCs were up to 100-fold more efficacious in EC cells compared to non-malignant cells and up to 200-fold more cytotoxic than drug treatment alone. Additionally, RAGE-targeting ADCs were not toxic in an in vivo pre-clinical mouse model, and significantly reduced tumour growth in a xenograft mouse model of disease. CONCLUSIONS: These data, together with important design considerations implied by the present study, suggest RAGE-ADCs could be translated to novel therapeutics for EC patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Immunoconjugates/therapeutic use , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Aged , Animals , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Biomarkers , Biomarkers, Tumor , Cell Line, Tumor , Disease Models, Animal , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Female , Gene Expression , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Immunoconjugates/pharmacokinetics , Immunohistochemistry , Mice , Middle Aged , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 24(10): 2452-2463, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29363524

ABSTRACT

Purpose: One third of ER-positive breast cancer patients who initially respond to endocrine therapy become resistant to treatment. Such treatment failure is associated with poor prognosis and remains an area of unmet clinical need. Here, we identify a specific posttranslational modification that occurs during endocrine resistance and which results in tumor susceptibility to the apoptosis-inducer TRAIL. This potentially offers a novel stratified approach to targeting endocrine-resistant breast cancer.Experimental Design: Cell line and primary-derived xenograft models of endocrine resistance were investigated for susceptibility to TRAIL. Tumor viability, cancer stem cell (CSC) viability (tumorspheres), tumor growth kinetics, and metastatic burden were assessed. Western blots for the TRAIL-pathway inhibitor, c-FLIP, and upstream regulators were performed. Results were confirmed in primary culture of 26 endocrine-resistant and endocrine-naïve breast tumors.Results: Breast cancer cell lines with acquired resistance to tamoxifen (TAMR) or faslodex were more sensitive to TRAIL than their endocrine-sensitive controls. Moreover, TRAIL eliminated CSC-like activity in TAMR cells, resulting in prolonged remission of xenografts in vivo In primary culture, TRAIL significantly depleted CSCs in 85% endocrine-resistant, compared with 8% endocrine-naïve, tumors, whereas systemic administration of TRAIL in endocrine-resistant patient-derived xenografts reduced tumor growth, CSC-like activity, and metastases. Acquired TRAIL sensitivity correlated with a reduction in intracellular levels of c-FLIP, and an increase in Jnk-mediated phosphorylation of E3-ligase, ITCH, which degrades c-FLIP.Conclusions: These results identify a novel mechanism of acquired vulnerability to an extrinsic cell death stimulus, in endocrine-resistant breast cancers, which has both therapeutic and prognostic potential. Clin Cancer Res; 24(10); 2452-63. ©2018 AACR.


Subject(s)
Breast Neoplasms/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Drug Resistance, Neoplasm , Protein Processing, Post-Translational , Receptors, Estrogen/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays
5.
Immunol Cell Biol ; 95(7): 620-629, 2017 08.
Article in English | MEDLINE | ID: mdl-28356569

ABSTRACT

The inherent resistance of cancer stem cells (CSCs) to existing therapies has largely hampered the development of effective treatments for advanced malignancy. To help develop novel immunotherapy approaches that efficiently target CSCs, an experimental model allowing reliable distinction of CSCs and non-CSCs was set up to study their interaction with non-MHC-restricted γδ T cells and antigen-specific CD8+ T cells. Stable lines with characteristics of breast CSC-like cells were generated from ras-transformed human mammary epithelial (HMLER) cells as confirmed by their CD44hi CD24lo GD2+ phenotype, their mesenchymal morphology in culture and their capacity to form mammospheres under non-adherent conditions, as well as their potent tumorigenicity, self-renewal and differentiation in xenografted mice. The resistance of CSC-like cells to γδ T cells could be overcome by inhibition of farnesyl pyrophosphate synthase (FPPS) through pretreatment with zoledronate or with FPPS-targeting short hairpin RNA. γδ T cells induced upregulation of MHC class I and CD54/ICAM-1 on CSC-like cells and thereby increased the susceptibility to antigen-specific killing by CD8+ T cells. Alternatively, γδ T-cell responses could be specifically directed against CSC-like cells using the humanised anti-GD2 monoclonal antibody hu14.18K322A. Our findings identify a powerful synergism between MHC-restricted and non-MHC-restricted T cells in the eradication of cancer cells including breast CSCs. Our research suggests that novel immunotherapies may benefit from a two-pronged approach combining γδ T-cell and CD8+ T-cell targeting strategies that triggers effective innate-like and tumour-specific adaptive responses.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , Antibodies/pharmacology , Breast/pathology , Cytotoxicity, Immunologic , Diphosphonates/pharmacology , Epithelial Cells/metabolism , Epitopes/immunology , Female , Humans , Imidazoles/pharmacology , Immunity, Innate , Interferon-gamma/metabolism , Major Histocompatibility Complex , Mice , Phenotype , Zoledronic Acid , ras Proteins/metabolism
7.
Cancer Lett ; 338(1): 57-62, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-22554712

ABSTRACT

Only a fraction of the cells in a breast tumour are able to seed new tumour growth. These so-called breast cancer stem cells (bCSCs) are characterised by a number of discrete functional properties, some of which impact on therapeutic strategies aimed at eliminating these cells from tumours. Here we discuss how recent experimental evidence indicates that phenotypic plasticity is a central feature of tumour cell heterogeneity and drug resistance, traits that must be overcome in order to efficiently target bCSCs as a therapy for breast cancer. We propose that a better understanding of this fundamental property of breast cancer stem cells, over and above their identification in tumours, is a priority for improvement of patient survival.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Breast Neoplasms/therapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Chemoradiotherapy , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Female , Humans , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects
8.
Breast Cancer Res ; 13(5): R88, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21914219

ABSTRACT

INTRODUCTION: It is postulated that breast cancer stem cells (bCSCs) mediate disease recurrence and drive formation of distant metastases - the principal cause of mortality in breast cancer patients. Therapeutic targeting of bCSCs, however, is hampered by their heterogeneity and resistance to existing therapeutics. In order to identify strategies to selectively remove bCSCs from breast cancers, irrespective of their clinical subtype, we sought an apoptosis mechanism that would target bCSCs yet would not kill normal cells. Suppression of the apoptosis inhibitor cellular FLICE-Like Inhibitory Protein (c-FLIP) partially sensitizes breast cancer cells to the anti-cancer agent Tumour Necrosis Factor-Related Apoptosis Inducing Ligand (TRAIL). Here we demonstrate in breast cancer cell lines that bCSCs are exquisitely sensitive to the de-repression of this pro-apoptotic pathway, resulting in a dramatic reduction in experimental metastases and the loss of bCSC self-renewal. METHODS: Suppression c-FLIP was performed by siRNA (FLIPi) in four breast cancer cell lines and by conditional gene-knockout in murine mammary glands. Sensitivity of these cells to TRAIL was determined by complementary cell apoptosis assays, including a novel heterotypic cell assay, while tumour-initiating potential of cancer stem cell subpopulations was determined by mammosphere cultures, aldefluor assay and in vivo transplantation. RESULTS: Genetic suppression of c-FLIP resulted in the partial sensitization of TRAIL-resistant cancer lines to the pro-apoptotic effects of TRAIL, irrespective of their cellular phenotype, yet normal mammary epithelial cells remained refractory to killing. While 10% to 30% of the cancer cell populations remained viable after TRAIL/FLIPi treatment, subsequent mammosphere and aldefluor assays demonstrated that this pro-apoptotic stimulus selectively targeted the functional bCSC pool, eliminating stem cell renewal. This culminated in an 80% reduction in primary tumours and a 98% reduction in metastases following transplantation. The recurrence of residual tumour initiating capacity was consistent with the observation that post-treated adherent cultures re-acquired bCSC-like properties in vitro. Importantly however this recurrent bCSC activity was attenuated following repeated TRAIL/FLIPi treatment. CONCLUSIONS: We describe an apoptotic mechanism that selectively and repeatedly removes bCSC activity from breast cancer cell lines and suggest that a combined TRAIL/FLIPi therapy could prevent metastatic disease progression in a broad range of breast cancer subtypes.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line, Tumor , Drug Screening Assays, Antitumor , Epithelium/metabolism , Epithelium/pathology , Estrogen Receptor alpha/metabolism , Female , Humans , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mice , Mice, Mutant Strains , RNA, Small Interfering , Receptor, ErbB-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...