Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667288

ABSTRACT

As the treatment landscape for prostate cancer gradually evolves, the frequency of treatment-induced neuroendocrine prostate cancer (NEPC) and double-negative prostate cancer (DNPC) that is deficient for androgen receptor (AR) and neuroendocrine (NE) markers has increased. These prostate cancer subtypes are typically refractory to AR-directed therapies and exhibit poor clinical outcomes. Only a small range of NEPC/DNPC models exist, limiting our molecular understanding of this disease and hindering our ability to perform preclinical trials exploring novel therapies to treat NEPC/DNPC that are urgently needed in the clinic. Here, we report the development of the CU-PC01 PDX model that represents AR-negative mCRPC with PTEN/RB/PSMA loss and CTNN1B/TP53/BRCA2 genetic variants. The CU-PC01 model lacks classic NE markers, with only focal and/or weak expression of chromogranin A, INSM1 and CD56. Collectively, these findings are most consistent with a DNPC phenotype. Ex vivo and in vivo preclinical studies revealed that CU-PC01 PDX tumours are resistant to mCRPC standard-of-care treatments enzalutamide and docetaxel, mirroring the donor patient's treatment response. Furthermore, short-term CU-PC01 tumour explant cultures indicate this model is initially sensitive to PARP inhibition with olaparib. Thus, the CU-PC01 PDX model provides a valuable opportunity to study AR-negative mCRPC biology and to discover new treatment avenues for this hard-to-treat disease.


Subject(s)
Piperazines , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , Xenograft Model Antitumor Assays , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Neoplasm Metastasis , Nitriles/pharmacology , Disease Models, Animal , Benzamides/pharmacology , Phthalazines/pharmacology , Phthalazines/therapeutic use
2.
Biomedicines ; 12(1)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38255248

ABSTRACT

The NF-κB co-factor Bcl3 is a proto-oncogene that promotes breast cancer proliferation, metastasis and therapeutic resistance, yet its role in breast cancer cell survival is unclear. Here, we sought to determine the effect of Bcl3 suppression alone on breast cancer cell viability, with a view to informing future studies that aim to target Bcl3 therapeutically. Bcl3 was suppressed by siRNA in breast cancer cell lines before changes in viability, proliferation, apoptosis and senescence were examined. Bcl3 suppression significantly reduced viability and was shown to induce apoptosis in all cell lines tested, while an additional p53-dependent senescence and senescence-associated secretory phenotype was also observed in those cells with functional p53. The role of the Bcl3/NF-κB axis in this senescence response was confirmed via siRNA of the non-canonical NF-κB subunit NFKB2/p52, which resulted in increased cellular senescence and the canonical subunit NFKB1/p50, which induced the senescence-associated secretory phenotype. An analysis of clinical data showed a correlation between reduced relapse-free survival in patients that expressed high levels of Bcl3 and carried a p53 mutation. Together, these data demonstrate a dual role for Bcl3/NF-κB in the maintenance of breast cancer cell viability and suggests that targeting Bcl3 may be more beneficial to patients with tumours that lack functional p53.

3.
Mol Cancer ; 23(1): 7, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195591

ABSTRACT

In the early 1990's a group of unrelated genes were identified from the sites of recurring translocations in B-cell lymphomas. Despite sharing the nomenclature 'Bcl', and an association with blood-borne cancer, these genes have unrelated functions. Of these genes, BCL2 is best known as a key cancer target involved in the regulation of caspases and other cell viability mechanisms. BCL3 on the other hand was originally identified as a non-canonical regulator of NF-kB transcription factor pathways - a signaling mechanism associated with important cell outcomes including many of the hallmarks of cancer. Most of the early investigations into BCL3 function have since focused on its role in NF-kB mediated cell proliferation, inflammation/immunity and cancer. However, recent evidence is coming to light that this protein directly interacts with and modulates a number of other signaling pathways including DNA damage repair, WNT/ß-catenin, AKT, TGFß/SMAD3 and STAT3 - all of which have key roles in cancer development, metastatic progression and treatment of solid tumours. Here we review the direct evidence demonstrating BCL3's central role in a transcriptional network of signaling pathways that modulate cancer biology and treatment response in a range of solid tumour types and propose common mechanisms of action of BCL3 which may be exploited in the future to target its oncogenic effects for patient benefit.


Subject(s)
Hematologic Neoplasms , NF-kappa B , Humans , Neoplasm Recurrence, Local , Proto-Oncogenes , Cell Proliferation
4.
Shoulder Elbow ; 14(2): 157-161, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35265181

ABSTRACT

Background: Pectoralis minor syndrome involves pain, paraesthesia and weakness in the arm due to compression of the brachial plexus passing beneath pectoralis minor; this paper reports the results of a single centre's treatment pathway in affected patients. Methods: During a four-year period, patients exhibiting symptoms of pectoralis minor syndrome without significant improvement following physiotherapy proceeded to Botulinum injection. Those with good response to injection but subsequent recurrence of symptoms were offered pectoralis minor tenotomy. Oxford shoulder Scores were collected at baseline and after interventions. Results: Twenty-one patients received Botulinum injection; at six weeks following injection, mean change in Oxford Shoulder Score was +12.4, with only one patient reporting a worsening of symptoms. Of the 17 patients with clinically significant response to injection, 12 have subsequently undergone tenotomy; three months following tenotomy, mean change in Oxford Shoulder Score from baseline was +22.3. Improvement was maintained in all patients at prolonged follow-up (average 20 months post-tenotomy). Discussion: This pathway has shown to be extremely effective in patients not responding to first-line treatment for pectoralis minor syndrome, with 85% of patients post-injection and 100% of patients post-tenotomy showing significant (greater than published minimal clinically important difference value of six points) improvements in Oxford Shoulder Score, maintained at follow-up.

5.
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
6.
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
7.
Int J Mol Sci ; 21(14)2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32679799

ABSTRACT

Proteins, as a major component of organisms, are considered the preferred biomaterials for drug delivery vehicles. Hemoglobin (Hb) has been recently rediscovered as a potential drug carrier, but its use for biomedical applications still lacks extensive investigation. To further explore the possibility of utilizing Hb as a potential tumor targeting drug carrier, we examined and compared the biodistribution of Hb in healthy and lung tumor-bearing mice, using for the first time 89Zr labelled Hb in a positron emission tomography (PET) measurement. Hb displays a very high conjugation yield in its fast and selective reaction with the maleimide-deferoxamine (DFO) bifunctional chelator. The high-resolution X-ray structure of the Hb-DFO complex demonstrated that cysteine ß93 is the sole attachment moiety to the αß-protomer of Hb. The Hb-DFO complex shows quantitative uptake of 89Zr in solution as determined by radiochromatography. Injection of 0.03 mg of Hb-DFO-89Zr complex in healthy mice indicates very high radioactivity in liver, followed by spleen and lungs, whereas a threefold increased dosage results in intensification of PET signal in kidneys and decreased signal in liver and spleen. No difference in biodistribution pattern is observed between naïve and tumor-bearing mice. Interestingly, the liver Hb uptake did not decrease upon clodronate-mediated macrophage depletion, indicating that other immune cells contribute to Hb clearance. This finding is of particular interest for rapidly developing clinical immunology and projects aiming to target, label or specifically deliver agents to immune cells.


Subject(s)
Drug Carriers/pharmacokinetics , Drug Delivery Systems , Hemoglobins/pharmacokinetics , Lung Neoplasms/metabolism , Lung/metabolism , Animals , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Deferoxamine/analogs & derivatives , Deferoxamine/pharmacokinetics , Drug Carriers/chemistry , Female , Hemoglobins/chemistry , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Positron Emission Tomography Computed Tomography , Radioisotopes/chemistry , Radioisotopes/pharmacokinetics , Tissue Distribution , Zirconium/chemistry , Zirconium/pharmacokinetics
8.
Carcinogenesis ; 41(10): 1432-1443, 2020 10 15.
Article in English | MEDLINE | ID: mdl-31957805

ABSTRACT

A key challenge in the implementation of anti-metastatics as cancer therapies is the multi-modal nature of cell migration, which allows tumour cells to evade the targeted inhibition of specific cell motility pathways. The nuclear factor-kappaB (NF-κB) co-factor B-cell lymphoma 3 (Bcl-3) has been implicated in breast cancer cell migration and metastasis, yet it remains to be determined exactly which cell motility pathways are controlled by Bcl-3 and whether migrating tumour cells are able to evade Bcl-3 intervention. Addressing these questions and the mechanism underpinning Bcl-3's role in this process would help determine its potential as a therapeutic target. Here we identify Bcl-3 as an upstream regulator of the two principal forms of breast cancer cell motility, involving collective and single-cell migration. This was found to be mediated by the master regulator Cdc42 through binding of the NF-κB transcription factor p50 to the Cdc42 promoter. Notably, Bcl-3 depletion inhibited both stable and transitory motility phenotypes in breast cancer cells with no evidence of migratory adaptation. Overexpression of Bcl-3 enhanced migration and increased metastatic tumour burden of breast cancer cells in vivo, whereas overexpression of a mutant Bcl-3 protein, which is unable to bind p50, suppressed cell migration and metastatic tumour burden suggesting that disruption of Bcl-3/NF-κB complexes is sufficient to inhibit metastasis. These findings identify a novel role for Bcl-3 in intrinsic and adaptive multi-modal cell migration mediated by its direct regulation of the Rho GTPase Cdc42 and identify the upstream Bcl-3:p50 transcription complex as a potential therapeutic target for metastatic disease.


Subject(s)
B-Cell Lymphoma 3 Protein/physiology , Breast Neoplasms/pathology , Cell Movement , NF-kappa B p50 Subunit/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , B-Cell Lymphoma 3 Protein/genetics , Cell Line, Tumor , Female , Humans , Mice , NF-kappa B p50 Subunit/genetics
9.
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
10.
Dis Model Mech ; 12(3)2019 03 04.
Article in English | MEDLINE | ID: mdl-30792270

ABSTRACT

To decrease bowel cancer incidence and improve survival, we need to understand the mechanisms that drive tumorigenesis. Recently, B-cell lymphoma 3 (BCL-3; a key regulator of NF-κB signalling) has been recognised as an important oncogenic player in solid tumours. Although reported to be overexpressed in a subset of colorectal cancers (CRCs), the role of BCL-3 expression in colorectal tumorigenesis remains poorly understood. Despite evidence in the literature that BCL-3 may interact with ß-catenin, it is perhaps surprising, given the importance of deregulated Wnt/ß-catenin/T-cell factor (TCF) signalling in colorectal carcinogenesis, that the functional significance of this interaction is not known. Here, we show for the first time that BCL-3 acts as a co-activator of ß-catenin/TCF-mediated transcriptional activity in CRC cell lines and that this interaction is important for Wnt-regulated intestinal stem cell gene expression. We demonstrate that targeting BCL-3 expression (using RNA interference) reduced ß-catenin/TCF-dependent transcription and the expression of intestinal stem cell genes LGR5 and ASCL2 In contrast, the expression of canonical Wnt targets Myc and cyclin D1 remained unchanged. Furthermore, we show that BCL-3 increases the functional stem cell phenotype, as shown by colorectal spheroid and tumoursphere formation in 3D culture conditions. We propose that BCL-3 acts as a driver of the stem cell phenotype in CRC cells, potentially promoting tumour cell plasticity and therapeutic resistance. As recent reports highlight the limitations of directly targeting cancer stem cells (CSCs), we believe that identifying and targeting drivers of stem cell plasticity have significant potential as new therapeutic targets.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , B-Cell Lymphoma 3 Protein , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Phenotype , Protein Binding , Protein Transport , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , TCF Transcription Factors/metabolism , Transcription Factors/genetics , beta Catenin/metabolism
11.
Cancer Discov ; 8(6): 764-779, 2018 06.
Article in English | MEDLINE | ID: mdl-29581176

ABSTRACT

Genetic alterations that potentiate PI3K signaling are frequent in prostate cancer, yet how different genetic drivers of the PI3K cascade contribute to prostate cancer is unclear. Here, we report PIK3CA mutation/amplification correlates with poor survival of patients with prostate cancer. To interrogate the requirement of different PI3K genetic drivers in prostate cancer, we employed a genetic approach to mutate Pik3ca in mouse prostate epithelium. We show Pik3caH1047R mutation causes p110α-dependent invasive prostate carcinoma in vivo Furthermore, we report that PIK3CA mutation and PTEN loss coexist in patients with prostate cancer and can cooperate in vivo to accelerate disease progression via AKT-mTORC1/2 hyperactivation. Contrasting single mutants that slowly acquire castration-resistant prostate cancer (CRPC), concomitant Pik3ca mutation and Pten loss caused de novo CRPC. Thus, Pik3ca mutation and Pten deletion are not functionally redundant. Our findings indicate that PIK3CA mutation is an attractive prognostic indicator for prostate cancer that may cooperate with PTEN loss to facilitate CRPC in patients.Significance: We show PIK3CA mutation correlates with poor prostate cancer prognosis and causes prostate cancer in mice. Moreover, PIK3CA mutation and PTEN loss coexist in prostate cancer and can cooperate in vivo to accelerate tumorigenesis and facilitate CRPC. Delineating this synergistic relationship may present new therapeutic/prognostic approaches to overcome castration/PI3K-AKT-mTORC1/2 inhibitor resistance. Cancer Discov; 8(6); 764-79. ©2018 AACR.See related commentary by Triscott and Rubin, p. 682This article is highlighted in the In This Issue feature, p. 663.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Mutation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Animals , Cell Line, Tumor , Disease Progression , Gene Amplification , Gene Deletion , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasms, Experimental , Prognosis , Survival Analysis
12.
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
13.
Technol Cancer Res Treat ; 16(6): 811-818, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28355964

ABSTRACT

PURPOSE: To assess the quality of cone beam computed tomography images obtained by a robotic arm-based and image-guided small animal conformal radiation therapy device. METHOD AND MATERIALS: The small animal conformal radiation therapy device is equipped with a 40 to 225 kV X-ray tube mounted on a custom made gantry, a 1024 × 1024 pixels flat panel detector (200 µm resolution), a programmable 6 degrees of freedom robot for cone beam computed tomography imaging and conformal delivery of radiation doses. A series of 2-dimensional radiographic projection images were recorded in cone beam mode by placing and rotating microcomputed tomography phantoms on the "palm' of the robotic arm. Reconstructed images were studied for image quality (spatial resolution, image uniformity, computed tomography number linearity, voxel noise, and artifacts). RESULTS: Geometric accuracy was measured to be 2% corresponding to 0.7 mm accuracy on a Shelley microcomputed tomography QA phantom. Qualitative resolution of reconstructed axial computed tomography slices using the resolution coils was within 200 µm. Quantitative spatial resolution was found to be 3.16 lp/mm. Uniformity of the system was measured within 34 Hounsfield unit on a QRM microcomputed tomography water phantom. Computed tomography numbers measured using the linearity plate were linear with material density (R2 > 0.995). Cone beam computed tomography images of the QRM multidisk phantom had minimal artifacts. CONCLUSION: Results showed that the small animal conformal radiation therapy device is capable of producing high-quality cone beam computed tomography images for precise and conformal small animal dose delivery. With its high-caliber imaging capabilities, the small animal conformal radiation therapy device is a powerful tool for small animal research.

14.
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
15.
Breast Cancer Res Treat ; 162(3): 465-477, 2017 04.
Article in English | MEDLINE | ID: mdl-28190248

ABSTRACT

PURPOSE: Nuclear factor-kappa B (NF-κB) signalling has been shown to regulate properties of breast cancer stem cells. However, the specific contribution of the non-canonical NF-κB pathway, components of which are elevated in aggressive breast cancer has not been addressed. METHODS: Through shRNA silencing of the Nfkb2 gene, the role of p100/p52 in 4T1 and N202.1A cell lines were assessed by NF-κB reporter, invasion, tumoursphere and orthotopic transplantation assays. The processing of p100 into p52 was also inhibited with a p97 ATPase inhibitor, NMS-873, and its effects on tumoursphere formation was assessed. RESULTS: Knockdown of Nfkb2 led to opposing changes in NF-κB-dependent transcription. NF-κB activity was elevated in 4T1 cells and this resulted in increased motility, cancer stem cell (CSC) activity and tumourigenicity in vivo. Conversely, depletion of Nfkb2 in N202.1a cells decreased NF-κB activity, CSC properties and tumourigenicity in vivo. By selectively overexpressing the p52 subunit in Nfkb2 depleted cells, we found that the increased malignancy in 4T1 cells could not be reverted in the presence of p52, whereas the decreased tumourigenicity of N202.1a cells could be rescued by p52. These results indicate that p100 and its subunit p52 have opposing effects on breast CSC activity. Accordingly, inhibition of an upstream regulator of p100 processing was effective in reducing tumoursphere formation of N202.1A and SKBR3 (ErbB2 HIGH) cells without aggravating that of 4T1 and MDA-MB-231 (ErbB2LOW) cells. CONCLUSION: These findings indicate that inhibiting the processing of p100 may be a potential therapeutic strategy to suppress CSC activity in a subset of breast tumours.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Neoplastic Stem Cells/metabolism , Acetanilides/pharmacology , Benzothiazoles/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Knockdown Techniques , Gene Silencing , Humans , Neoplastic Stem Cells/pathology
16.
Technol Cancer Res Treat ; 16(1): 45-56, 2017 02.
Article in English | MEDLINE | ID: mdl-26792490

ABSTRACT

We have developed a small animal conformal radiation therapy device that provides a degree of geometrical/anatomical targeting comparable to what is achievable in a commercial animal irradiator. small animal conformal radiation therapy device is capable of producing precise and accurate conformal delivery of radiation to target as well as for imaging small animals. The small animal conformal radiation therapy device uses an X-ray tube, a robotic animal position system, and a digital imager. The system is in a steel enclosure with adequate lead shielding following National Council on Radiation Protection and Measurements 49 guidelines and verified with Geiger-Mueller survey meter. The X-ray source is calibrated following AAPM TG-61 specifications and mounted at 101.6 cm from the floor, which is a primary barrier. The X-ray tube is mounted on a custom-made "gantry" and has a special collimating assembly system that allows field size between 0.5 mm and 20 cm at isocenter. Three-dimensional imaging can be performed to aid target localization using the same X-ray source at custom settings and an in-house reconstruction software. The small animal conformal radiation therapy device thus provides an excellent integrated system to promote translational research in radiation oncology in an academic laboratory. The purpose of this article is to review shielding and dosimetric measurement and highlight a few successful studies that have been performed to date with our system. In addition, an example of new data from an in vivo rat model of breast cancer is presented in which spatially fractionated radiation alone and in combination with thermal ablation was applied and the therapeutic benefit examined.


Subject(s)
Radiotherapy, Conformal/instrumentation , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/radiotherapy , Cone-Beam Computed Tomography/instrumentation , Cone-Beam Computed Tomography/methods , Dose Fractionation, Radiation , Female , Radiation Protection , Radiometry , Radiotherapy Dosage , Radiotherapy, Conformal/methods , Radiotherapy, Image-Guided/instrumentation , Radiotherapy, Image-Guided/methods , Rats , X-Rays
17.
Phytother Res ; 30(3): 418-25, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26666387

ABSTRACT

The essential oils from Commiphora species have for centuries been recognized to possess medicinal properties. Here, we performed gas chromatography-mass spectrometry on the essential oil from opoponax (Commiphora guidotti) and identified bisabolene isomers as the main constituents of this essential oil. Opoponax essential oil, a chemical component; ß-bisabolene and an alcoholic analogue, α-bisabolol, were tested for their ability to selectively kill breast cancer cells. Only ß-bisabolene, a sesquiterpene constituting 5% of the essential oil, exhibited selective cytotoxic activity for mouse cells (IC50 in normal Eph4: >200 µg/ml, MG1361: 65.49 µg/ml, 4T1: 48.99 µg/ml) and human breast cancer cells (IC50 in normal MCF-10A: 114.3 µg/ml, MCF-7: 66.91 µg/ml, MDA-MB-231: 98.39 µg/ml, SKBR3: 70.62 µg/ml and BT474: 74.3 µg/ml). This loss of viability was because of the induction of apoptosis as shown by Annexin V-propidium iodide and caspase-3/7 activity assay. ß-bisabolene was also effective in reducing the growth of transplanted 4T1 mammary tumours in vivo (37.5% reduction in volume by endpoint). In summary, we have identified an anti-cancer agent from the essential oil of opoponax that exhibits specific cytotoxicity to both human and murine mammary tumour cells in vitro and in vivo, and this warrants further investigation into the use of ß-bisabolene in the treatment of breast cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Commiphora/chemistry , Oils, Volatile/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Sesquiterpenes/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Female , Gas Chromatography-Mass Spectrometry , Humans , MCF-7 Cells , Mice , Monocyclic Sesquiterpenes , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Plant Extracts/pharmacology , Sesquiterpenes/pharmacology
18.
Mol Cancer ; 14: 209, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26667821

ABSTRACT

BACKGROUND: The clinical application of TRAIL receptor agonists as a novel cancer therapy has been tempered by heterogeneity in tumour responses. This is illustrated in breast cancer, where TRAIL is cytotoxic in cell lines of mesenchymal origin but refractory in lines with an epithelial-like phenotype. However, it is now evident that intra-tumour heterogeneity includes a minority subpopulation of tumour-initiating stem/progenitor-like cells (CSCs) that possess mesenchymal characteristics. We hypothesised therefore that TRAIL may target these phenotypically distinct CSC-like cells that are common to most - if not all - breast cancers, thus impacting on the source of malignancy in a much broader range of breast tumour subtypes than previously envisaged. METHODS: We used colony formation, tumoursphere, flow cytometry and xenograft tumour initiation assays to observe the TRAIL sensitivity of CSC-like cells in a panel of two mesenchymal-like (TRAIL-sensitive) and four epithelial-like (TRAIL-resistant) breast cancer cell lines. Subcellular levels of the endogenous TRAIL inhibitor, cFLIP, were determined by western blot and immunofluorescence microscopy. The effect of the subcellular redistribution of cFLIP on TRAIL sensitivity and Wnt signalling was determined using cFLIP localisation mutants and the TOPFlash reporter assay respectively. RESULTS: TRAIL universally suppressed the clonal expansion of stem/progenitors in all six of the breast cancer cell lines tested, irrespective of their phenotype or overall sensitivity to TRAIL. A concomitant reduction in tumour initiation was confirmed in the TRAIL-resistant epithelial cell line, MCF-7, following serial dilution xenotransplantation. Furthermore TRAIL sensitivity of breast CSCs was inversely proportional to the relative cytoplasmic levels of cFLIP while overexpression of cFLIP in the cytosol using subcellular localization mutants of cFLIP protected these cells from cytotoxicity. The accumulation of nuclear cFLIP on the other hand did not influence TRAIL cytotoxicity but instead promoted Wnt-dependent signalling. CONCLUSION: These data propose a novel role for TRAIL as a selective CSC agent with a broad specificity for both epithelial and mesenchymal breast tumour subtypes. Furthermore we identify a dual role for cFLIP in the maintenance of breast CSC viability, dependent upon its subcellular distribution.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Neoplastic Stem Cells/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cytoplasm/metabolism , Drug Resistance, Neoplasm , Female , Humans , Mice, Nude , Neoplastic Stem Cells/physiology , Protein Transport , Wnt Signaling Pathway , Xenograft Model Antitumor Assays
20.
PLoS Biol ; 12(2): e1001799, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586114

ABSTRACT

Developmental programming links growth in early life with health status in adulthood. Although environmental factors such as maternal diet can influence the growth and adult health status of offspring, the genetic influences on this process are poorly understood. Using the mouse as a model, we identify the imprinted gene Grb10 as a mediator of nutrient supply and demand in the postnatal period. The combined actions of Grb10 expressed in the mother, controlling supply, and Grb10 expressed in the offspring, controlling demand, jointly regulate offspring growth. Furthermore, Grb10 determines the proportions of lean and fat tissue during development, thereby influencing energy homeostasis in the adult. Most strikingly, we show that the development of normal lean/fat proportions depends on the combined effects of Grb10 expressed in the mother, which has the greater effect on offspring adiposity, and Grb10 expressed in the offspring, which influences lean mass. These distinct functions of Grb10 in mother and pup act complementarily, which is consistent with a coadaptation model of imprinting evolution, a model predicted but for which there is limited experimental evidence. In addition, our findings identify Grb10 as a key genetic component of developmental programming, and highlight the need for a better understanding of mother-offspring interactions at the genetic level in predicting adult disease risk.


Subject(s)
Body Size/genetics , GRB10 Adaptor Protein/genetics , Animals , Female , GRB10 Adaptor Protein/metabolism , Gene Expression Regulation, Developmental , Genomic Imprinting , Karyopherins/physiology , Lactation/genetics , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/physiology , STAT5 Transcription Factor/physiology , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...