Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Int J Cancer ; 144(6): 1356-1366, 2019 03 15.
Article En | MEDLINE | ID: mdl-30125350

Renal cell cancer (RCC) has become a prototype example of the extensive intratumor heterogeneity and clonal evolution of human cancers. However, there is little direct evidence on how the genetic heterogeneity impacts on drug response profiles of the cancer cells. Our goal was to determine how genomic clonal evolution impacts drug responses. Finding from our study could help to define the challenge that clonal evolution poses on cancer therapy. We established multiple patient-derived cells (PDCs) from different tumor regions of four RCC patients, verified their clonal relationship to each other and to the uncultured tumor tissue by genome sequencing. Furthermore, comprehensive drug-sensitivity testing with 460 oncological drugs was performed on all PDC clones. The PDCs retained many cancer-specific copy number alterations and mutations in driver genes such as VHL, PBRM1, PIK3C2A, KMD5C and TSC2 genes. The drug testing highlighted vulnerability in the PDCs toward approved RCC drugs, such as the mTOR-inhibitor temsirolimus, but also novel sensitivities were uncovered. The individual PDC clones from different tumor regions in a patient showed distinct drug-response profiles, suggesting that genomic heterogeneity contributes to the variability in drug responses. Studies of multiple PDCs from a patient with cancer are informative for elucidating cancer heterogeneity and for the determination on how the genomic evolution is manifested in cancer drug responsiveness. This approach could facilitate tailoring of drugs and drug combinations to individual patients.


Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Clonal Evolution , Drug Resistance, Neoplasm/genetics , Kidney Neoplasms/drug therapy , 3T3 Cells , Adult , Aged , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Coculture Techniques , DNA Copy Number Variations , Drug Screening Assays, Antitumor/methods , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mice , Middle Aged , Mutation , Primary Cell Culture , Tumor Cells, Cultured
2.
J Pathol ; 245(1): 101-113, 2018 05.
Article En | MEDLINE | ID: mdl-29443392

A key question in precision medicine is how functional heterogeneity in solid tumours informs therapeutic sensitivity. We demonstrate that spatial characteristics of oncogenic signalling and therapy response can be modelled in precision-cut slices from Kras-driven non-small-cell lung cancer with varying histopathologies. Unexpectedly, profiling of in situ tumours demonstrated that signalling stratifies mostly according to histopathology, showing enhanced AKT and SRC activity in adenosquamous carcinoma, and mitogen-activated protein kinase (MAPK) activity in adenocarcinoma. In addition, high intertumour and intratumour variability was detected, particularly of MAPK and mammalian target of rapamycin (mTOR) complex 1 activity. Using short-term treatment of slice explants, we showed that cytotoxic responses to combination MAPK and phosphoinositide 3-kinase-mTOR inhibition correlate with the spatially defined activities of both pathways. Thus, whereas genetic drivers determine histopathology spectra, histopathology-associated and spatially variable signalling activities determine drug sensitivity. Our study is in support of spatial aspects of signalling heterogeneity being considered in clinical diagnostic settings, particularly to guide the selection of drug combinations. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Carcinogenesis/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mitogen-Activated Protein Kinases/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology
3.
Eur Urol ; 71(3): 319-327, 2017 03.
Article En | MEDLINE | ID: mdl-27160946

BACKGROUND: Technology development to enable the culture of human prostate cancer (PCa) progenitor cells is required for the identification of new, potentially curative therapies for PCa. OBJECTIVE: We established and characterized patient-derived conditionally reprogrammed cells (CRCs) to assess their biological properties and to apply these to test the efficacies of drugs. DESIGN, SETTING, AND PARTICIPANTS: CRCs were established from seven patient samples with disease ranging from primary PCa to advanced castration-resistant PCa (CRPC). The CRCs were characterized by genomic, transcriptomic, protein expression, and drug profiling. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The phenotypic quantification of the CRCs was done based on immunostaining followed by image analysis with Advanced Cell Classifier using Random Forest supervised machine learning. Copy number aberrations (CNAs) were called from whole-exome sequencing and transcriptomics using in-house pipelines. Dose-response measurements were used to generate multiparameter drug sensitivity scores using R-statistical language. RESULTS AND LIMITATIONS: We generated six benign CRC cultures which all had an androgen receptor-negative, basal/transit-amplifying phenotype with few CNAs. In three-dimensional cell culture, these cells could re-express the androgen receptor. The CRCs from a CRPC patient (HUB.5) displayed multiple CNAs, many of which were shared with the parental tumor. We carried out high-throughput drug-response studies with 306 emerging and clinical cancer drugs. Using the benign CRCs as controls, we identified the Bcl-2 family inhibitor navitoclax as the most potent cancer-specific drug for the CRCs from a CRPC patient. Other drug efficacies included taxanes, mepacrine, and retinoids. CONCLUSIONS: Comprehensive cancer pharmacopeia-wide drug testing of CRCs from a CRPC patient highlighted both known and novel drug sensitivities in PCa, including navitoclax, which is currently being tested in clinical trials of CRPC. PATIENT SUMMARY: We describe an approach to generate patient-derived cancer cells from advanced prostate cancer and apply such cells to discover drugs that could be applied in clinical trials for castration-resistant prostate cancer.


Antineoplastic Agents/pharmacology , Cellular Reprogramming Techniques , Precision Medicine , Prostatic Neoplasms, Castration-Resistant/drug therapy , Tumor Cells, Cultured/drug effects , Aniline Compounds/pharmacology , Bexarotene , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Kallikreins/metabolism , Keratin-18/metabolism , Keratin-5/metabolism , Male , Organoplatinum Compounds/pharmacology , Oxaliplatin , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Quinacrine/pharmacology , Receptors, Androgen/metabolism , Sulfonamides/pharmacology , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology
4.
Sci Rep ; 5: 17187, 2015 Dec 09.
Article En | MEDLINE | ID: mdl-26647838

Precision-cut slices of in vivo tumours permit interrogation in vitro of heterogeneous cells from solid tumours together with their native microenvironment. They offer a low throughput but high content in vitro experimental platform. Using mouse models as surrogates for three common human solid tumours, we describe a standardised workflow for systematic comparison of tumour slice cultivation methods and a tissue microarray-based method to archive them. Cultivated slices were compared to their in vivo source tissue using immunohistochemical and transcriptional biomarkers, particularly of cellular stress. Mechanical slicing induced minimal stress. Cultivation of tumour slices required organotypic support materials and atmospheric oxygen for maintenance of integrity and was associated with significant temporal and loco-regional changes in protein expression, for example HIF-1α. We recommend adherence to the robust workflow described, with recognition of temporal-spatial changes in protein expression before interrogation of tumour slices by pharmacological or other means.


Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Animals , Biomarkers , Cell Line, Tumor , Gene Expression , Heterografts , Humans , Immunohistochemistry/methods , Mice , Oxygen/metabolism , Principal Component Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction , Stress, Physiological , Tissue Array Analysis , Tissue Culture Techniques
5.
EMBO Mol Med ; 7(8): 994-5, 2015 Aug.
Article En | MEDLINE | ID: mdl-26034096

The availability of blood-based markers to predict response of a solid tumor to treatment, estimate patient prognosis and diagnose relapse well before clinical symptoms arise, is a long-standing hope in clinical oncology. Ideally, assays designed to provide such information should be inexpensive (at least in the foreseeable future), simple, and, of course, predictive of the clinical evolution of the disease. While early research focused on circulating glycosylated tumor-derived protein biomarkers, the focus is now rapidly shifting to new opportunities, such as circulating tumor cells, extracellular vesicles, micro-RNAs and cancer-derived cell-free DNA a.k.a. circulating tumor-derived DNA (ctDNA).


Biomarkers, Tumor/blood , Breast Neoplasms/complications , Breast Neoplasms/pathology , DNA/blood , Neoplasm Metastasis/diagnosis , Female , Humans
6.
J Urol ; 191(3): 842-9, 2014 Mar.
Article En | MEDLINE | ID: mdl-24035881

PURPOSE: DNA damage responses are relevant to prostate cancer initiation, progression and treatment. Few models of the normal and malignant human prostate that maintain stromal-epithelial interactions in vivo exist in which to study DNA damage responses. We evaluated the feasibility of maintaining tissue slice grafts at subcutaneous vs subrenal capsular sites in RAG2(-/-)γC(-/-) mice to study the DNA damage responses of normal and malignant glands. MATERIALS AND METHODS: We compared the take rate and histology of tissue slice grafts from fresh, precision cut surgical specimens that were maintained for 1 to 4 weeks in subcutaneous vs subrenal capsular sites. Induction of γH2AX, p53, ATM and apoptosis was evaluated as a measure of the DNA damage response after irradiation. RESULTS: The take rate of subcutaneous tissue slice grafts was higher than typically reported but lower than at the subrenal capsular site. Subcutaneous tissue slice grafts frequently showed basal cell hyperplasia, squamous metaplasia and cystic atrophy, and cancer did not survive. In contrast, normal and malignant histology was well maintained in subrenal capsular tissue slice grafts. Regardless of implantation site the induction of γH2AX and ATM occurred in tissue slice graft epithelium 1 hour after irradiation and decreased to basal level by 24 hours, indicating DNA damage recognition and repair. As observed previously in prostatic ex vivo models, p53 was not activated. Notably, tumor but not normal cells responded to irradiation by undergoing apoptosis. CONCLUSIONS: To our knowledge this is the first study of DNA damage responses in a patient derived prostate tissue graft model. The subrenal capsular site of RAG2(-/-)γC(-/-) mice optimally maintains normal and malignant histology and function, permitting novel studies of DNA damage responses in a physiological context.


DNA Damage , Prostatic Neoplasms/genetics , Tissue Transplantation/methods , Animals , Apoptosis , DNA Repair , Humans , Immunohistochemistry , Male , Mice , Microscopy, Fluorescence , Neoplasm Grading , Prostate/pathology , Prostatic Neoplasms/pathology
7.
Prostate ; 73(2): 219-26, 2013 Jan.
Article En | MEDLINE | ID: mdl-22806587

BACKGROUND: PSA is the most useful prostate cancer marker. However, its levels are increased also in some non-malignant conditions. In circulation, the majority of PSA is complexed with protease inhibitors, including α(1) -antichymotrypsin (ACT). The proportion of the PSA-ACT complex is higher in patients with prostate cancer than in controls without cancer. The expression of ACT has been shown to be higher in prostate cancer than in benign prostatic hyperplasia. However, results regarding the extent which PSA forms complexes within the prostate and whether there are differences in complex formation between normal and malignant prostatic tissue are inconsistent and limited. METHODS: We studied complex formation of PSA secreted by cultured human prostate tissues and in the tissue by in situ proximity ligation assay (PLA). Free, total and active PSA, and the PSA-ACT complex were determined in tissue culture media by immunoassays, immunoblotting, and chromatographic methods. RESULTS: The majority of PSA in tissue culture medium was free and enzymatically active. However, a significant proportion (1.6 ± 0.5%) of immunoreactive PSA was found to be complexed with ACT. Complex formation was confirmed by in situ PLA, which showed more intense staining of PSA-ACT in cancers with Gleason grade 3 than in adjacent benign tissues from the same patients. CONCLUSIONS: These results show that PSA forms complexes already within the prostate and that PSA-ACT levels are increased in moderately differentiated prostate cancer tissue. This may explain, at least partially, why the ratio of serum PSA-ACT to total PSA is increased in prostate cancer.


Prostate-Specific Antigen/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , alpha 1-Antichymotrypsin/metabolism , Adult , Aged , Cell Differentiation/physiology , Humans , Male , Middle Aged , Multiprotein Complexes/metabolism , Prostate/pathology , Prostatic Neoplasms/diagnosis , Protein Binding/physiology
8.
Prostate ; 72(10): 1060-70, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22072329

BACKGROUND: Prostate and seminal vesicle are two similar hormone responsive human organs that differ dramatically in their cancer incidence. DNA damage response (DDR) is required for maintenance of genomic integrity. METHODS: In this study we investigated the DDR and cell cycle checkpoint activation of these organs using orthotopic cultures of human surgery-derived tissues and primary cultures of isolated prostate and seminal vesicle cells. RESULTS: We find that the activation of ATM signaling pathway by ionizing radiation (IR) was comparable in both tissues. Previously, we have shown that the prostate secretory cells express low levels of histone variant H2AX and phosphorylated H2AX (γH2AX) after IR. Here we demonstrate that H2AX levels are low also in the secretory seminal vesicle cells suggesting that this is a common phenotype of postmitotic cells. We consequently established primary epithelial cell cultures from both organs to compare their DDR. Interestingly, contrary to human prostate epithelial cells (HPEC), primary seminal vesicle epithelial cells (HSVEC) displayed effective cell cycle checkpoints after IR and expressed higher levels of Wee1A checkpoint kinase. Furthermore, HSVEC but not HPEC cells were able to activate p53 and to induce p21 cell cycle inhibitor. DISCUSSION: Our results show that during replication, the checkpoint enforcement is more proficient in the seminal vesicle than in the prostate epithelium cells. This indicates a more stringent enforcement of DDR in replicating seminal vesicle epithelial cells, and suggests that epithelial regeneration combined with sub-optimal checkpoint responses may contribute to high frequency of genetic lesions in the prostate epithelium.


Cell Cycle Checkpoints/genetics , DNA Damage/genetics , Epithelial Cells/physiology , Prostate/physiology , Seminal Vesicles/physiology , Cells, Cultured , Epithelial Cells/pathology , Epithelium/pathology , Epithelium/physiology , Humans , Male , Prostate/pathology , Seminal Vesicles/pathology
9.
Cell Cycle ; 10(20): 3545-53, 2011 Oct 15.
Article En | MEDLINE | ID: mdl-22030624

The ability of cells to respond and repair DNA damage is fundamental for the maintenance of genomic integrity. Ex vivo culturing of surgery-derived human tissues has provided a significant advancement to assess DNA damage response (DDR) in the context of normal cytoarchitecture in a non-proliferating tissue. Here, we assess the dependency of prostate epithelium DDR on ATM and DNA-PKcs, the major kinases responsible for damage detection and repair by nonhomologous end-joining (NHEJ), respectively. DNA damage was caused by ionizing radiation (IR) and cytotoxic drugs, cultured tissues were treated with ATM and DNA-PK inhibitors, and DDR was assessed by phosphorylation of ATM and its targets H2AX and KAP1, a heterochromatin binding protein. Phosphorylation of H2AX and KAP1 was fast, transient and fully dependent on ATM, but these responses were moderate in luminal cells. In contrast, DNA-PKcs was phosphorylated in both luminal and basal cells, suggesting that DNA-PK-dependent repair was also activated in the luminal cells despite the diminished H2AX and KAP1 responses. These results indicate that prostate epithelial cell types have constitutively dissimilar responses to DNA damage. We correlate the altered damage response to the differential chromatin state of the cells. These findings are relevant in understanding how the epithelium senses and responds to DNA damage.


Cell Cycle Proteins/metabolism , DNA Damage , DNA End-Joining Repair/physiology , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Epithelium/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Histones/metabolism , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation , Prostate/cytology , Radiation, Ionizing , Repressor Proteins/metabolism , Tripartite Motif-Containing Protein 28
10.
PLoS One ; 5(9): e12996, 2010 Sep 27.
Article En | MEDLINE | ID: mdl-20885994

Manipulation of the activity of the p53 tumor suppressor pathway has demonstrated potential benefit in preclinical mouse tumor models and has entered human clinical trials. We describe here an improved, extensive small-molecule chemical compound library screen for p53 pathway activation in a human cancer cell line devised to identify hits with potent antitumor activity. We uncover six novel small-molecule lead compounds, which activate p53 and repress the growth of human cancer cells. Two tested compounds suppress in vivo tumor growth in an orthotopic mouse model of human B-cell lymphoma. All compounds interact with DNA, and two activate p53 pathway in a DNA damage signaling-dependent manner. A further screen of a drug library of approved drugs for medicinal uses and analysis of gene-expression signatures of the novel compounds revealed similarities to known DNA intercalating and topoisomerase interfering agents and unexpected connectivities to known drugs without previously demonstrated anticancer activities. These included several neuroleptics, glycosides, antihistamines and adrenoreceptor antagonists. This unbiased screen pinpoints interference with the DNA topology as the predominant mean of pharmacological activation of the p53 pathway and identifies potential novel antitumor agents.


Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Neoplasms/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/physiopathology , Small Molecule Libraries/therapeutic use , Tumor Suppressor Protein p53/genetics
11.
Cancer Res ; 70(21): 8630-41, 2010 Nov 01.
Article En | MEDLINE | ID: mdl-20978201

DNA damage response (DDR) pathways have been extensively studied in cancer cell lines and mouse models, but little is known about how DNA damage is recognized by different cell types in nonmalignant, slowly replicating human tissues. Here, we assess, using ex vivo cultures of human prostate tissue, DDR caused by cytotoxic drugs (camptothecin, doxorubicin, etoposide, and cisplatin) and ionizing radiation (IR) in the context of normal tissue architecture. Using specific markers for basal and luminal epithelial cells, we determine and quantify cell compartment-specific damage recognition. IR, doxorubicin, and etoposide induced the phosphorylation of H2A.X on Ser(139) (γH2AX) and DNA damage foci formation. Surprisingly, luminal epithelial cells lack the prominent γH2AX response after IR when compared with basal cells, although ATM phosphorylation on Ser(1981) and 53BP1 foci were clearly detectable in both cell types. The attenuated γH2AX response seems to result from low levels of total H2A.X in the luminal cells. Marked increase in p53, a downstream target of the activated ATM pathway, was detected only in response to camptothecin and doxorubicin. These findings emphasize the diversity of pathways activated by DNA damage in slowly replicating tissues and reveal an unexpected deviation in the prostate luminal compartment that may be relevant in prostate tumorigenesis. Detailed mapping of tissue and cell type differences in DDR will provide an outlook of relevant responses to therapeutic strategies.


Cell Cycle Proteins/metabolism , DNA Damage/physiology , DNA-Binding Proteins/metabolism , Prostate/metabolism , Prostate/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , DNA Damage/radiation effects , Histones/metabolism , Humans , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins , Male , Phosphorylation/radiation effects , Radiation, Ionizing , Tumor Suppressor p53-Binding Protein 1
12.
Proc Natl Acad Sci U S A ; 104(17): 7211-6, 2007 Apr 24.
Article En | MEDLINE | ID: mdl-17431037

Cellular DNA damage triggers the DNA damage response pathway and leads to enforcement of cell cycle checkpoints, which are essential for the maintenance of genomic integrity and are activated in early stages of tumorigenesis. A special feature of prostate cancer is its high incidence and multifocality. To address the functionality of DNA damage checkpoints in the prostate, we analyzed the responses of human primary prostate epithelial cells (HPECs) and freshly isolated human prostate tissues to gamma-irradiation. We find that gamma-irradiation activates the ataxia telangiectasia mutated-associated DNA damage response pathway in the HPECs but that the clearance of phosphorylated histone H2AX (gammaH2AX) foci is delayed. Surprisingly, gamma-irradiated HPECs were unable to enforce cell cycle checkpoint arrest and had sustained cyclin-dependent kinase 2 (Cdk2)-associated kinase activity because of a lack of inhibitory Cdk phosphorylation by Wee1A tyrosine kinase. We further show that HPECs express low levels of Wee1A and that ectopic Wee1A efficiently rescues the checkpoints. We recapitulate the absence of checkpoint responses in epithelium of ex vivo irradiated human prostate tissue despite robust induction of gammaH2AX. The findings show that prostate epithelium has a surprising inability to control checkpoint arrest, the lack of which may predispose to accrual of DNA lesions.


Cell Cycle Proteins/metabolism , Cell Cycle , DNA Damage , Epithelium/enzymology , Nuclear Proteins/metabolism , Prostate/enzymology , Protein-Tyrosine Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/radiation effects , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation/radiation effects , Epithelial Cells/cytology , Epithelial Cells/enzymology , Epithelial Cells/radiation effects , Epithelium/radiation effects , Humans , Male , Phosphotyrosine/metabolism , Prostate/cytology , Prostate/radiation effects , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
...