Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nat Commun ; 15(1): 4653, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821942

ABSTRACT

Patient-derived xenograft (PDX) models are widely used in cancer research. To investigate the genomic fidelity of non-small cell lung cancer PDX models, we established 48 PDX models from 22 patients enrolled in the TRACERx study. Multi-region tumor sampling increased successful PDX engraftment and most models were histologically similar to their parent tumor. Whole-exome sequencing enabled comparison of tumors and PDX models and we provide an adapted mouse reference genome for improved removal of NOD scid gamma (NSG) mouse-derived reads from sequencing data. PDX model establishment caused a genomic bottleneck, with models often representing a single tumor subclone. While distinct tumor subclones were represented in independent models from the same tumor, individual PDX models did not fully recapitulate intratumor heterogeneity. On-going genomic evolution in mice contributed modestly to the genomic distance between tumors and PDX models. Our study highlights the importance of considering primary tumor heterogeneity when using PDX models and emphasizes the benefit of comprehensive tumor sampling.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Genetic Heterogeneity , Lung Neoplasms , Mice, Inbred NOD , Mice, SCID , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Animals , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Female , Exome Sequencing , Genomics/methods , Male , Xenograft Model Antitumor Assays , Heterografts , Disease Models, Animal , Aged , Middle Aged
2.
Mol Ther ; 32(5): 1497-1509, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38429928

ABSTRACT

The hallmark of epidermolysis bullosa (EB) is fragile attachment of epithelia due to genetic variants in cell adhesion genes. We describe 16 EB patients treated in the ear, nose, and throat department of a tertiary pediatric hospital linked to the United Kingdom's national EB unit between 1992 and 2023. Patients suffered a high degree of morbidity and mortality from laryngotracheal stenosis. Variants in laminin subunit alpha-3 (LAMA3) were found in 10/15 patients where genotype was available. LAMA3 encodes a subunit of the laminin-332 heterotrimeric extracellular matrix protein complex and is expressed by airway epithelial basal stem cells. We investigated the benefit of restoring wild-type LAMA3 expression in primary EB patient-derived basal cell cultures. EB basal cells demonstrated weak adhesion to cell culture substrates, but could otherwise be expanded similarly to non-EB basal cells. In vitro lentiviral overexpression of LAMA3A in EB basal cells enabled them to differentiate in air-liquid interface cultures, producing cilia with normal ciliary beat frequency. Moreover, transduction restored cell adhesion to levels comparable to a non-EB donor culture. These data provide proof of concept for a combined cell and gene therapy approach to treat airway disease in LAMA3-affected EB.


Subject(s)
Cell Adhesion , Epidermolysis Bullosa , Laminin , Lentivirus , Humans , Laminin/metabolism , Laminin/genetics , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Epidermolysis Bullosa/therapy , Epidermolysis Bullosa/pathology , Child , Lentivirus/genetics , Male , Female , Child, Preschool , Genetic Therapy/methods , Genetic Vectors/genetics , Epithelial Cells/metabolism , Cells, Cultured , Gene Expression , Adolescent , Infant
3.
Cell Rep Med ; 4(2): 100915, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36657447

ABSTRACT

More than half of patients with malignant mesothelioma show alterations in the BAP1 tumor-suppressor gene. Being a member of the Polycomb repressive deubiquitinating (PR-DUB) complex, BAP1 loss results in an altered epigenome, which may create new vulnerabilities that remain largely unknown. Here, we performed a CRISPR-Cas9 kinome screen in mesothelioma cells that identified two kinases in the mevalonate/cholesterol biosynthesis pathway. Furthermore, our analysis of chromatin, expression, and genetic perturbation data in mesothelioma cells suggests a dependency on PR complex 2 (PRC2)-mediated silencing. Pharmacological inhibition of PRC2 elevates the expression of cholesterol biosynthesis genes only in BAP1-deficient mesothelioma, thereby sensitizing these cells to the combined targeting of PRC2 and the mevalonate pathway. Finally, by subjecting autochthonous Bap1-deficient mesothelioma mice or xenografts to mevalonate pathway inhibition (zoledronic acid) and PRC2 inhibition (tazemetostat), we demonstrate a potent anti-tumor effect, suggesting a targeted combination therapy for Bap1-deficient mesothelioma.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Humans , Animals , Mice , Mevalonic Acid , Lung Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Mesothelioma/genetics , Mesothelioma/pathology , Cholesterol , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
4.
J Biol Chem ; 297(5): 101223, 2021 11.
Article in English | MEDLINE | ID: mdl-34597666

ABSTRACT

Malignant pleural mesothelioma (MPM) is a rare, aggressive, and incurable cancer arising from the mesothelial lining of the pleura, with few available treatment options. We recently reported that loss of function of the nuclear deubiquitinase BRCA1-associated protein 1 (BAP1), a frequent event in MPM, is associated with sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis. As a potential underlying mechanism, here we report that BAP1 negatively regulates the expression of TRAIL receptors: death receptor 4 (DR4) and death receptor 5 (DR5). Using tissue microarrays of tumor samples from MPM patients, we found a strong inverse correlation between BAP1 and TRAIL receptor expression. BAP1 knockdown increased DR4 and DR5 expression, whereas overexpression of BAP1 had the opposite effect. Reporter assays confirmed wt-BAP1, but not catalytically inactive BAP1 mutant, reduced promoter activities of DR4 and DR5, suggesting deubiquitinase activity is required for the regulation of gene expression. Co-immunoprecipitation studies demonstrated direct binding of BAP1 to the transcription factor Ying Yang 1 (YY1), and chromatin immunoprecipitation assays revealed BAP1 and YY1 to be enriched in the promoter regions of DR4 and DR5. Knockdown of YY1 also increased DR4 and DR5 expression and sensitivity to TRAIL. These results suggest that BAP1 and YY1 cooperatively repress transcription of TRAIL receptors. Our finding that BAP1 directly regulates the extrinsic apoptotic pathway will provide new insights into the role of BAP1 in the development of MPM and other cancers with frequent BAP1 mutations.


Subject(s)
Mesothelioma, Malignant/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , YY1 Transcription Factor/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mesothelioma, Malignant/genetics , Mutation , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , YY1 Transcription Factor/genetics
5.
Stem Cell Res Ther ; 11(1): 256, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32586403

ABSTRACT

BACKGROUND: MSCTRAIL is a cell-based therapy consisting of human allogeneic umbilical cord-derived MSCs genetically modified to express the anti-cancer protein TRAIL. Though cell-based therapies are typically designed with a target tissue in mind, delivery is rarely assessed due to a lack of translatable non-invasive imaging approaches. In this preclinical study, we demonstrate 89Zr-oxine labelling and PET-CT imaging as a potential clinical solution for non-invasively tracking MSCTRAIL biodistribution. Future implementation of this technique should improve our understanding of MSCTRAIL during its evaluation as a therapy for metastatic lung adenocarcinoma. METHODS: MSCTRAIL were radiolabelled with 89Zr-oxine and assayed for viability, phenotype, and therapeutic efficacy post-labelling. PET-CT imaging of 89Zr-oxine-labelled MSCTRAIL was performed in a mouse model of lung cancer following intravenous injection, and biodistribution was confirmed ex vivo. RESULTS: MSCTRAIL retained the therapeutic efficacy and MSC phenotype in vitro at labelling amounts up to and above those required for clinical imaging. The effect of 89Zr-oxine labelling on cell proliferation rate was amount- and time-dependent. PET-CT imaging showed delivery of MSCTRAIL to the lungs in a mouse model of lung cancer up to 1 week post-injection, validated by in vivo bioluminescence imaging, autoradiography, and fluorescence imaging on tissue sections. CONCLUSIONS: 89Zr-oxine labelling and PET-CT imaging present a potential method of evaluating the biodistribution of new cell therapies in patients, including MSCTRAIL. This offers to improve understanding of cell therapies, including mechanism of action, migration dynamics, and inter-patient variability.


Subject(s)
Lung Neoplasms , Positron Emission Tomography Computed Tomography , Humans , Lung , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/therapy , Oxyquinoline , Tissue Distribution
6.
Oncol Rep ; 42(1): 35-42, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115546

ABSTRACT

Drug-resistant neuroblastoma remains a major challenge in paediatric oncology and novel and less toxic therapeutic approaches are urgently needed to improve survival and reduce the side effects of traditional therapeutic interventions. Mesenchymal stem cells (MSCs) are an attractive candidate for cell and gene therapy since they are recruited by and able to infiltrate tumours. This feature has been exploited by creating genetically modified MSCs that are able to combat cancer by delivering therapeutic molecules. Whether neuroblastomas attract systemically delivered MSCs is still controversial. We investigated whether MSCs engineered to express tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) could: i) cause death of classic and primary neuroblastoma cell lines in vitro; ii) migrate to tumour sites in vivo; and iii) reduce neuroblastoma growth in xenotransplantation experiments. We observed that classic and primary neuroblastoma cell lines expressing death receptors could be killed by TRAIL-loaded MSCs in vitro. When injected in the peritoneum of neuroblastoma-bearing mice, TRAIL-MSCs migrated to tumour sites, but were unable to change the course of cancer development. These results indicated that MSCs have the potential to be used to deliver drugs in neuroblastoma patients, but more effective biopharmaceuticals should be used instead of TRAIL.


Subject(s)
Genetic Engineering/methods , Mesenchymal Stem Cells/cytology , Neuroblastoma/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Female , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Neuroblastoma/metabolism , Neuroblastoma/therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays
7.
Tissue Eng Part C Methods ; 25(2): 103-113, 2019 02.
Article in English | MEDLINE | ID: mdl-30648471

ABSTRACT

IMPACT STATEMENT: Methodologies for incorporation of cells into tissue-engineered grafts, particularly at the later preclinical stages, are suboptimal and non-validated, and monitoring cell fate within scaffolds cultured in bioreactors and in vivo is challenging. In this study, we demonstrate how bioluminescence imaging (BLI) can overcome these difficulties and allow quantitative cell tracking at multiple stages of the bioengineering preclinical pipeline. Our robust bioluminescence-based approach allowed reproducible longitudinal monitoring of mesoangioblast localization and survival in 2D/3D tissue culture, in organ-scale bioreactors, and in vivo. Our findings will encourage the use of BLI in tissue engineering studies, improving the overall quality of cell-scaffold interaction research.


Subject(s)
Bioengineering/methods , Cell Tracking/methods , Esophagus/physiology , Luminescent Measurements/methods , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/cytology , Myoblasts/cytology , Cell Differentiation , Cells, Cultured , Child , Humans , Image Processing, Computer-Assisted , Myoblasts/transplantation , Tissue Scaffolds
8.
Lung Cancer ; 127: 164-166, 2019 01.
Article in English | MEDLINE | ID: mdl-30642545

ABSTRACT

Introduction BRCA1 associated protein-1 (BAP1) is a key tumor driver in mesothelioma and a potential biomarker predicting response to several targeted therapies in clinical testing. Whether it also modulates response to cytotoxic chemotherapy is undetermined. This study used retrospective response analysis of BAP1 expression in archival tumor biopsies taken from patients in the MS01 trial (NCT00075699). We aimed to determine if BAP1 expression correlated with overall survival within the three treatment arms in this trial, namely active symptom control (ASC); ASC plus mitomycin, vinblastine and cisplatin (MVP); and ASC plus vinorelbine. Materials and methods We used immunohistochemical analysis of tumor samples from the MS01 trial to identify subgroups with and without nuclear BAP1 expression. We performed correlative analysis of clinical characteristics (age at diagnosis, sex and histological subtype) and overall survival within treatment arms with nuclear BAP1 expression. Results 89 tumor samples from the 409 patients originally in the trial were available for analysis. Of these, 60 samples harbored a positive internal control, in the form of positive staining of inflammatory cells for BAP1, and were carried forward for analysis. Correlative analysis suggested no significant association between loss of nuclear BAP1 expression and age at diagnosis, sex and histological subtype. Kaplan Meier survival analysis revealed a small, though non-significant, overall survival disadvantage associated with BAP1 expression in tumors from patients treated with vinorelbine. Discussion This exploratory analysis suggests BAP1 expression may modify response to vinorelbine in MPM, possibly due to prevention of mitotic microtubule formation. We suggest ongoing and planned clinical studies of vinorelbine in MPM assess BAP1 expression as a predictive biomarker of response.


Subject(s)
Biomarkers, Tumor/metabolism , Cisplatin/therapeutic use , Mesothelioma/diagnosis , Pleural Neoplasms/diagnosis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Vinblastine/therapeutic use , Aged , Female , Humans , Male , Mesothelioma/drug therapy , Mesothelioma/mortality , Microtubules/metabolism , Pleural Neoplasms/drug therapy , Pleural Neoplasms/mortality , Prognosis , Retrospective Studies , Survival Analysis
9.
Elife ; 72018 01 18.
Article in English | MEDLINE | ID: mdl-29345617

ABSTRACT

Malignant mesothelioma (MM) is poorly responsive to systemic cytotoxic chemotherapy and invariably fatal. Here we describe a screen of 94 drugs in 15 exome-sequenced MM lines and the discovery of a subset defined by loss of function of the nuclear deubiquitinase BRCA associated protein-1 (BAP1) that demonstrate heightened sensitivity to TRAIL (tumour necrosis factor-related apoptosis-inducing ligand). This association is observed across human early passage MM cultures, mouse xenografts and human tumour explants. We demonstrate that BAP1 deubiquitinase activity and its association with ASXL1 to form the Polycomb repressive deubiquitinase complex (PR-DUB) impacts TRAIL sensitivity implicating transcriptional modulation as an underlying mechanism. Death receptor agonists are well-tolerated anti-cancer agents demonstrating limited therapeutic benefit in trials without a targeting biomarker. We identify BAP1 loss-of-function mutations, which are frequent in MM, as a potential genomic stratification tool for TRAIL sensitivity with immediate and actionable therapeutic implications.


Subject(s)
Lung Neoplasms/physiopathology , Mesothelioma/physiopathology , Repressor Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line, Tumor , Humans , Mesothelioma, Malignant , Mice
10.
Clin Cancer Res ; 24(1): 84-94, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29061644

ABSTRACT

Purpose: Despite intense research, treatment options for patients with mesothelioma are limited and offer only modest survival advantage. We screened a large panel of compounds in multiple mesothelioma models and correlated sensitivity with a range of molecular features to detect biomarkers of drug response.Experimental design: We utilized a high-throughput chemical inhibitor screen in a panel of 889 cancer cell lines, including both immortalized and primary early-passage mesothelioma lines, alongside comprehensive molecular characterization using Illumina whole-exome sequencing, copy-number analysis and Affymetrix array whole transcriptome profiling. Subsequent validation was done using functional assays such as siRNA silencing and mesothelioma mouse xenograft models.Results: A subgroup of immortalized and primary MPM lines appeared highly sensitive to FGFR inhibition. None of these lines harbored genomic alterations of FGFR family members, but rather BAP1 protein loss was associated with enhanced sensitivity to FGFR inhibition. This was confirmed in an MPM mouse xenograft model and by BAP1 knockdown and overexpression in cell line models. Gene expression analyses revealed an association between BAP1 loss and increased expression of the receptors FGFR1/3 and ligands FGF9/18. BAP1 loss was associated with activation of MAPK signaling. These associations were confirmed in a cohort of MPM patient samples.Conclusions: A subgroup of mesotheliomas cell lines harbor sensitivity to FGFR inhibition. BAP1 protein loss enriches for this subgroup and could serve as a potential biomarker to select patients for FGFR inhibitor treatment. These data identify a clinically relevant MPM subgroup for consideration of FGFR therapeutics in future clinical studies. Clin Cancer Res; 24(1); 84-94. ©2017 AACR.


Subject(s)
Gene Expression Profiling , Lung Neoplasms/genetics , Mesothelioma/genetics , Pharmacogenetics , Pleural Neoplasms/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , Female , Fibroblast Growth Factors/metabolism , Gene Amplification , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mesothelioma/drug therapy , Mesothelioma/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Pharmacogenetics/methods , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , RNA Interference , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Xenograft Model Antitumor Assays
11.
J Extracell Vesicles ; 6(1): 1265291, 2017.
Article in English | MEDLINE | ID: mdl-28326166

ABSTRACT

Extracellular vesicles (EVs) are lipid membrane-enclosed nanoparticles released by cells. They mediate intercellular communication by transferring biological molecules and therefore have potential as innovative drug delivery vehicles. TNF-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis of cancer cells. Unfortunately, the clinical application of recombinant rTRAIL has been hampered by its low bioavailability and resistance of cancer cells. EV-mediated TRAIL delivery may circumvent these problems. Mesenchymal stromal cells (MSCs) produce EVs and could be a good source for therapeutic EV production. We investigated if TRAIL could be expressed in MSC-derived EVs and examined their cancer cell-killing efficacy. EVs were isolated by ultracentrifugation and were membranous particles of 50-70 nm in diameter. Both MSC- and TRAIL-expressing MSC (MSCT)-derived EVs express CD63, CD9 and CD81, but only MSCT-EVs express surface TRAIL. MSCT-EVs induced apoptosis in 11 cancer cell lines in a dose-dependent manner but showed no cytotoxicity in primary human bronchial epithelial cells. Caspase activity inhibition or TRAIL neutralisation blocked the cytotoxicity of TRAIL-positive EVs. MSCT-EVs induced pronounced apoptosis in TRAIL-resistant cancer cells and this effect could be further enhanced using a CDK9 inhibitor. These data indicate that TRAIL delivery by MSC-derived EVs is an effective anticancer therapy.

12.
Cytotherapy ; 18(7): 860-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27260207

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) are being extensively researched for cell therapy and tissue engineering. We have engineered MSCs to express the pro-apoptotic protein tumor necrosis factor-related apoptosis inducing ligand (TRAIL) and are currently preparing this genetically modified cell therapy for a phase 1/2a clinical trial in patients with metastatic lung cancer. To do this, we need to prepare a cryopreserved allogeneic MSCTRAIL cell bank for further expansion before patient delivery. The effects of cryopreservation on a genetically modified cell therapy product have not been clearly determined. METHODS: We tested different concentrations of dimethyl sulfoxide (DMSO) added to the human serum albumin ZENALB 4.5 and measured post-thaw cell viability, proliferation ability and differentiation characteristics. In addition, we examined the homing ability, TRAIL expression and cancer cell-killing capacities of cryopreserved genetically modified MSCs compared with fresh, continually cultured cells. RESULTS: We demonstrated that the post-thaw viability of MSCs in 5% DMSO (v/v) with 95% ZENALB 4.5 (v/v) is 85.7 ± 0.4%, which is comparable to that in conventional freezing media. We show that cryopreservation does not affect the long-term expression of TRAIL and that cryopreserved TRAIL-expressing MSCs exhibit similar levels of homing and, importantly, retain their potency in triggering cancer cell death. CONCLUSIONS: This study shows that cryopreservation is unlikely to affect the therapeutic properties of MSCTRAIL and supports the generation of a cryopreserved master cell bank.


Subject(s)
Cryopreservation/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Shape , Cell Survival , Cells, Cultured , Freezing , Humans , Neoplasms/metabolism , Neoplasms/pathology , Receptors, CXCR4/metabolism
13.
Sci Rep ; 6: 24006, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27035095

ABSTRACT

Although squamous cell carcinomas (SqCCs) of the lungs, head and neck, oesophagus, and cervix account for up to 30% of cancer deaths, the mechanisms that regulate disease progression remain incompletely understood. Here, we use gene transduction and human tumor xenograft assays to establish that the tumour suppressor Cell adhesion molecule 1 (CADM1) inhibits SqCC proliferation and invasion, processes fundamental to disease progression. We determine that the extracellular domain of CADM1 mediates these effects by forming a complex with HER2 and integrin α6ß4 at the cell surface that disrupts downstream STAT3 activity. We subsequently show that treating CADM1 null tumours with the JAK/STAT inhibitor ruxolitinib mimics CADM1 gene restoration in preventing SqCC growth and metastases. Overall, this study identifies a novel mechanism by which CADM1 prevents SqCC progression and suggests that screening tumours for loss of CADM1 expression will help identify those patients most likely to benefit from JAK/STAT targeted chemotherapies.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Expression Profiling , Humans , Immunoglobulins/genetics , Integrin alpha6beta4/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Nitriles , Pyrazoles/chemistry , Pyrimidines , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
14.
Cytotherapy ; 17(7): 885-96, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25888191

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cell (MSC) delivery of pro-apoptotic tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is an attractive strategy for anticancer therapy. MSCs expressing full-length human TRAIL (flT) or its soluble form (sT) have previously been shown to be effective for cancer killing. However, a comparison between the two forms has never been performed, leaving it unclear which approach is most effective. This study addresses the issue for the possible clinical application of TRAIL-expressing MSCs in the future. METHODS: MSCs were transduced with lentiviruses expressing flT or an isoleucine zipper-fused sT. TRAIL expression was examined and cancer cell apoptosis was measured after treatment with transduced MSCs or with MSC-derived soluble TRAIL. RESULTS: The transduction does not adversely affect cell phenotype. The sT-transduced MSCs (MSC-sT) secrete abundant levels of soluble TRAIL but do not present the protein on the cell surface. Interestingly, the flT-transduced MSCs (MSC-flT) not only express cell-surface TRAIL but also release flT into medium. These cells were examined for inducing apoptosis in 20 cancer cell lines. MSC-sT cells showed very limited effects. By contrast, MSC-flT cells demonstrated high cancer cell-killing efficiency. More importantly, MSC-flT cells can overcome some cancer cell resistance to recombinant TRAIL. In addition, both cell surface flT and secreted flT are functional for inducing apoptosis. The secreted flT was found to have higher cancer cell-killing capacity than either recombinant TRAIL or MSC-secreted sT. CONCLUSIONS: These observations demonstrate that MSC delivery of flT is superior to MSC delivery of sT for cancer therapy.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/metabolism , Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Mesenchymal Stem Cells/cytology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transduction, Genetic/methods
15.
Thorax ; 69(7): 638-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24567297

ABSTRACT

Malignant pleural mesothelioma is a rare but devastating cancer of the pleural lining with no effective treatment. The tumour is often diffusely spread throughout the chest cavity, making surgical resection difficult, while systemic chemotherapy offers limited benefit. Bone marrow-derived mesenchymal stem cells (MSCs) home to and incorporate into tumour stroma, making them good candidates to deliver anticancer therapies. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a pro-apoptotic molecule that selectively induces apoptosis in cancer cells, leaving healthy cells unaffected. We hypothesised that human MSCs expressing TRAIL (MSCTRAIL) would home to an in vivo model of malignant pleural mesothelioma and reduce tumour growth. Human MSCs transduced with a lentiviral vector encoding TRAIL were shown in vitro to kill multiple malignant mesothelioma cell lines as predicted by sensitivity to recombinant TRAIL (rTRAIL). In vivo MSC homing was delineated using dual fluorescence and bioluminescent imaging, and we observed that higher levels of MSC engraftment occur after intravenous delivery compared with intrapleural delivery of MSCs. Finally, we show that intravenous delivery of MSCTRAIL results in a reduction in malignant pleural mesothelioma tumour growth in vivo via an increase in tumour cell apoptosis.


Subject(s)
Lung Neoplasms/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Administration, Topical , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Immunohistochemistry , Infusions, Intravenous , Lung Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Mice, Inbred NOD , Mice, SCID , Pleural Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transfection , Tumor Burden/drug effects , Tumor Cells, Cultured
16.
Respiration ; 85(6): 443-51, 2013.
Article in English | MEDLINE | ID: mdl-23711430

ABSTRACT

Despite recent advances in treatment, lung cancer accounts for one third of all cancer-related deaths, underlining the need of development of new therapies. Mesenchymal stem cells (MSCs) possess the ability to specifically home into tumours and their metastases. This property of MSCs could be exploited for the delivery of various anti-tumour agents directly into tumours. However, MSCs are not simple delivery vehicles but cells with active physiological process. This review outlines various agents which can be delivered by MSCs with substantial emphasis on TRAIL (tumour necrosis factor-related apoptosis-inducing ligand).


Subject(s)
Lung Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , Humans
17.
J Pathol ; 229(4): 608-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23208928

ABSTRACT

Epidermal growth factor receptor (EGFR) pathway activation is a frequent event in human carcinomas. Mutations in EGFR itself are, however, rare, and the mechanisms regulating EGFR activation remain elusive. Leucine-rich immunoglobulin repeats-1 (LRIG1), an inhibitor of EGFR activity, is one of four genes identified that predict patient survival across solid tumour types including breast, lung, melanoma, glioma, and bladder. We show that deletion of Lrig1 is sufficient to promote murine airway hyperplasia through loss of contact inhibition and that re-expression of LRIG1 in human lung cancer cells inhibits tumourigenesis. LRIG1 regulation of contact inhibition occurs via ternary complex formation with EGFR and E-cadherin with downstream modulation of EGFR activity. We find that LRIG1 LOH is frequent across cancers and its loss is an early event in the development of human squamous carcinomas. Our findings imply that the early stages of squamous carcinoma development are driven by a change in amplitude of EGFR signalling governed by the loss of contact inhibition.


Subject(s)
Cadherins/metabolism , Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Precancerous Conditions/genetics , Animals , Cadherins/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Contact Inhibition , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Loss of Heterozygosity , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiprotein Complexes , Nerve Tissue Proteins/metabolism , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Sequence Deletion , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...