Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Death Dis ; 15(1): 32, 2024 01 11.
Article in English | MEDLINE | ID: mdl-38212297

ABSTRACT

Immune checkpoint blockade (ICB) provides effective and durable responses for several tumour types by unleashing an immune response directed against cancer cells. However, a substantial number of patients treated with ICB develop relapse or do not respond, which has been partly attributed to the immune-suppressive effect of tumour hypoxia. We have previously demonstrated that the mitochondrial complex III inhibitor atovaquone alleviates tumour hypoxia both in human xenografts and in cancer patients by decreasing oxygen consumption and consequently increasing oxygen availability in the tumour. Here, we show that atovaquone alleviates hypoxia and synergises with the ICB antibody anti-PD-L1, significantly improving the rates of tumour eradication in the syngeneic CT26 model of colorectal cancer. The synergistic effect between atovaquone and anti-PD-L1 relied on CD8+ T cells, resulted in the establishment of a tumour-specific memory immune response, and was not associated with any toxicity. We also tested atovaquone in combination with anti-PD-L1 in the LLC (lung) and MC38 (colorectal) cancer syngeneic models but, despite causing a considerable reduction in tumour hypoxia, atovaquone did not add any therapeutic benefit to ICB in these models. These results suggest that atovaquone has the potential to improve the outcomes of patients treated with ICB, but predictive biomarkers are required to identify individuals likely to benefit from this intervention.


Subject(s)
Electron Transport Complex III , Neoplasms , Humans , Animals , Mice , Atovaquone/pharmacology , Atovaquone/therapeutic use , Neoplasms/drug therapy , CD8-Positive T-Lymphocytes , Immunotherapy/methods , B7-H1 Antigen , Tumor Microenvironment
2.
Clin Cancer Res ; 29(8): 1631-1642, 2023 04 14.
Article in English | MEDLINE | ID: mdl-36689546

ABSTRACT

PURPOSE: DNA polymerase theta (Polθ, encoded by the POLQ gene) is a DNA repair enzyme critical for microhomology mediated end joining (MMEJ). Polθ has limited expression in normal tissues but is frequently overexpressed in cancer cells and, therefore, represents an ideal target for tumor-specific radiosensitization. In this study we evaluate whether targeting Polθ with novel small-molecule inhibitors is a feasible strategy to improve the efficacy of radiotherapy. EXPERIMENTAL DESIGN: We characterized the response to Polθ inhibition in combination with ionizing radiation in different cancer cell models in vitro and in vivo. RESULTS: Here, we show that ART558 and ART899, two novel and specific allosteric inhibitors of the Polθ DNA polymerase domain, potently radiosensitize tumor cells, particularly when combined with fractionated radiation. Importantly, noncancerous cells were not radiosensitized by Polθ inhibition. Mechanistically, we show that the radiosensitization caused by Polθ inhibition is most effective in replicating cells and is due to impaired DNA damage repair. We also show that radiosensitization is still effective under hypoxia, suggesting that these inhibitors may help overcome hypoxia-induced radioresistance. In addition, we describe for the first time ART899 and characterize it as a potent and specific Polθ inhibitor with improved metabolic stability. In vivo, the combination of Polθ inhibition using ART899 with fractionated radiation is well tolerated and results in a significant reduction in tumor growth compared with radiation alone. CONCLUSIONS: These results pave the way for future clinical trials of Polθ inhibitors in combination with radiotherapy.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , Cell Line, Tumor
3.
STAR Protoc ; 3(2): 101355, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35542177

ABSTRACT

The colony formation assay is the gold-standard technique to assess cell viability after treatment with cytotoxic reagents, ionizing radiation, and cytotoxic combinatorial treatments. This protocol describes a high-throughput automated and high-content imaging approach to screen siRNA molecular libraries in HeLa cervical cancer cells in 96-well format. We detail reverse transfection of cells with siRNAs, followed by ionizing radiation, fixing, and staining of the plates for automated colony counting. This protocol can be used across a broad range of cell types. For complete details on the use and execution of this protocol, please refer to Tiwana et al. (2015).


Subject(s)
High-Throughput Screening Assays , Radiation, Ionizing , Gene Library , High-Throughput Screening Assays/methods , RNA, Small Interfering/genetics , Transfection
4.
Nat Commun ; 12(1): 3636, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140467

ABSTRACT

To identify approaches to target DNA repair vulnerabilities in cancer, we discovered nanomolar potent, selective, low molecular weight (MW), allosteric inhibitors of the polymerase function of DNA polymerase Polθ, including ART558. ART558 inhibits the major Polθ-mediated DNA repair process, Theta-Mediated End Joining, without targeting Non-Homologous End Joining. In addition, ART558 elicits DNA damage and synthetic lethality in BRCA1- or BRCA2-mutant tumour cells and enhances the effects of a PARP inhibitor. Genetic perturbation screening revealed that defects in the 53BP1/Shieldin complex, which cause PARP inhibitor resistance, result in in vitro and in vivo sensitivity to small molecule Polθ polymerase inhibitors. Mechanistically, ART558 increases biomarkers of single-stranded DNA and synthetic lethality in 53BP1-defective cells whilst the inhibition of DNA nucleases that promote end-resection reversed these effects, implicating these in the synthetic lethal mechanism-of-action. Taken together, these observations describe a drug class that elicits BRCA-gene synthetic lethality and PARP inhibitor synergy, as well as targeting a biomarker-defined mechanism of PARPi-resistance.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , DNA Repair/drug effects , DNA-Directed DNA Polymerase/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Synthetic Lethal Mutations/drug effects , Allosteric Regulation , Animals , Apoptosis/drug effects , Apoptosis/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Deoxyribonucleases/antagonists & inhibitors , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Homologous Recombination/drug effects , Humans , Inhibitory Concentration 50 , Mice , Organoids/drug effects , Ovarian Neoplasms/genetics , Rats , Synthetic Lethal Mutations/genetics , Tumor Suppressor p53-Binding Protein 1/deficiency , Tumor Suppressor p53-Binding Protein 1/metabolism , DNA Polymerase theta
5.
Clin Cancer Res ; 27(9): 2459-2469, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33597271

ABSTRACT

PURPOSE: Tumor hypoxia fuels an aggressive tumor phenotype and confers resistance to anticancer treatments. We conducted a clinical trial to determine whether the antimalarial drug atovaquone, a known mitochondrial inhibitor, reduces hypoxia in non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: Patients with NSCLC scheduled for surgery were recruited sequentially into two cohorts: cohort 1 received oral atovaquone at the standard clinical dose of 750 mg twice daily, while cohort 2 did not. Primary imaging endpoint was change in tumor hypoxic volume (HV) measured by hypoxia PET-CT. Intercohort comparison of hypoxia gene expression signatures using RNA sequencing from resected tumors was performed. RESULTS: Thirty patients were evaluable for hypoxia PET-CT analysis, 15 per cohort. Median treatment duration was 12 days. Eleven (73.3%) atovaquone-treated patients had meaningful HV reduction, with median change -28% [95% confidence interval (CI), -58.2 to -4.4]. In contrast, median change in untreated patients was +15.5% (95% CI, -6.5 to 35.5). Linear regression estimated the expected mean HV was 55% (95% CI, 24%-74%) lower in cohort 1 compared with cohort 2 (P = 0.004), adjusting for cohort, tumor volume, and baseline HV. A key pharmacodynamics endpoint was reduction in hypoxia-regulated genes, which were significantly downregulated in atovaquone-treated tumors. Data from multiple additional measures of tumor hypoxia and perfusion are presented. No atovaquone-related adverse events were reported. CONCLUSIONS: This is the first clinical evidence that targeting tumor mitochondrial metabolism can reduce hypoxia and produce relevant antitumor effects at the mRNA level. Repurposing atovaquone for this purpose may improve treatment outcomes for NSCLC.


Subject(s)
Atovaquone/pharmacology , Gene Expression Regulation, Neoplastic , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics , Atovaquone/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Energy Metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Molecular Imaging , Positron Emission Tomography Computed Tomography , STAT3 Transcription Factor/metabolism
6.
Cell Death Differ ; 28(4): 1333-1346, 2021 04.
Article in English | MEDLINE | ID: mdl-33168956

ABSTRACT

T-LAK-originated protein kinase (TOPK) overexpression is a feature of multiple cancers, yet is absent from most phenotypically normal tissues. As such, TOPK expression profiling and the development of TOPK-targeting pharmaceutical agents have raised hopes for its future potential in the development of targeted therapeutics. Results presented in this paper confirm the value of TOPK as a potential target for the treatment of solid tumours, and demonstrate the efficacy of a TOPK inhibitor (OTS964) when used in combination with radiation treatment. Using H460 and Calu-6 lung cancer xenograft models, we show that pharmaceutical inhibition of TOPK potentiates the efficacy of fractionated irradiation. Furthermore, we provide in vitro evidence that TOPK plays a hitherto unknown role during S phase, showing that TOPK depletion increases fork stalling and collapse under conditions of replication stress and exogenous DNA damage. Transient knockdown of TOPK was shown to impair recovery from fork stalling and to increase the formation of replication-associated single-stranded DNA foci in H460 lung cancer cells. We also show that TOPK interacts directly with CHK1 and Cdc25c, two key players in the checkpoint signalling pathway activated after replication fork collapse. This study thus provides novel insights into the mechanism by which TOPK activity supports the survival of cancer cells, facilitating checkpoint signalling in response to replication stress and DNA damage.


Subject(s)
Checkpoint Kinase 1/drug effects , Lung Neoplasms/radiotherapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Radiation Tolerance/drug effects , cdc25 Phosphatases/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/radiation effects , Female , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Radiation Tolerance/genetics , Signal Transduction , Survival Rate , Xenograft Model Antitumor Assays , cdc25 Phosphatases/genetics , cdc25 Phosphatases/radiation effects
7.
Cell Death Discov ; 6: 110, 2020.
Article in English | MEDLINE | ID: mdl-33133645

ABSTRACT

Platinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malarial agent by demonstrating that it potentiates cancer cell death of a subset of platinums. We show that ATQ in combination with carboplatin or cisplatin induces striking and repeatable concentration- and time-dependent cell death sensitization in vitro across a variety of cancer cell lines. ATQ induces mitochondrial reactive oxygen species (mROS), depleting intracellular glutathione (GSH) pools in a concentration-dependent manner. The superoxide dismutase mimetic MnTBAP rescues ATQ-induced mROS production and pre-loading cells with the GSH prodrug N-acetyl cysteine (NAC) abrogates the sensitization. Together, these findings implicate ATQ-induced oxidative stress as key mediator of the sensitizing effect. At physiologically achievable concentrations, ATQ and carboplatin furthermore synergistically delay the growth of three-dimensional avascular spheroids. Clinically, ATQ is a safe and specific inhibitor of the electron transport chain (ETC) and is concurrently being repurposed as a candidate tumor hypoxia modifier. Together, these findings suggest that ATQ is deserving of further study as a candidate platinum sensitizing agent.

8.
Cancers (Basel) ; 12(4)2020 Mar 26.
Article in English | MEDLINE | ID: mdl-32224979

ABSTRACT

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

9.
Prostate ; 79(10): 1079-1089, 2019 07.
Article in English | MEDLINE | ID: mdl-31104332

ABSTRACT

BACKGROUND: Enhancer of zeste 2 (EZH2) promotes prostate cancer progression. We hypothesized that increased EZH2 expression is associated with postradiotherapy metastatic disease recurrence, and may promote radioresistance. METHODS: EZH2 expression was investigated using immunohistochemistry in diagnostic prostate biopsies of 113 prostate cancer patients treated with radiotherapy with curative intent. Associations between EZH2 expression in malignant and benign tissue in prostate biopsy cores and outcomes were investigated using univariate and multivariate Cox regression analyses. LNCaP and PC3 cell radiosensitivity was investigated using colony formation and γH2AX assays following UNC1999 chemical probe-mediated EZH2 inhibition. RESULTS: While there was no significant association between EZH2 expression and biochemical recurrence following radiotherapy, univariate analysis revealed that prostate cancer cytoplasmic and total EZH2 expression were significantly associated with metastasis development postradiotherapy (P = 0.034 and P = 0.003, respectively). On multivariate analysis, the prostate cancer total EZH2 expression score remained statistically significant (P = 0.003), while cytoplasmic EZH2 expression did not reach statistical significance (P = 0.053). No association was observed between normal adjacent prostate EZH2 expression and biochemical recurrence or metastasis. LNCaP and PC3 cell treatment with UNC1999 reduced histone H3 lysine 27 tri-methylation levels. Irradiation of LNCaP or PC3 cells with a single 2 Gy fraction with UNC1999-mediated EZH2 inhibition resulted in a statistically significant, though modest, reduction in cell colony number for both cell lines. Increased γH2AX foci were observed 24 hours after ionizing irradiation in LNCaP cells, but not in PC3, following UNC1999-mediated EZH2 inhibition vs controls. CONCLUSIONS: Taken together, these results reveal that high pretreatment EZH2 expression in prostate cancer in diagnostic biopsies is associated with an increased risk of postradiotherapy metastatic disease recurrence, but EZH2 function may only at most play a modest role in promoting prostate cancer cell radioresistance.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Disease Progression , Humans , Male , Neoplasm Grading , Prostate/pathology , Prostatic Neoplasms/radiotherapy , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/secondary
10.
Eur J Cancer ; 113: 87-95, 2019 05.
Article in English | MEDLINE | ID: mdl-30991262

ABSTRACT

BACKGROUND: Pre-clinically, phosphoinositide 3-kinase (PI3K) inhibition radiosensitises tumours by increasing intrinsic radiosensitivity and by reducing tumour hypoxia. We assessed whether buparlisib, a class 1 PI3K inhibitor, can be safely combined with radiotherapy in patients with non-small cell lung carcinoma (NSCLC) and investigated its effect on tumour hypoxia. METHODS: This was a 3 + 3 dose escalation and dose expansion phase I trial in patients with advanced NSCLC. Buparlisib dose levels were 50 mg, 80 mg and 100 mg once daily orally for 2 weeks, with palliative thoracic radiotherapy (20 Gy in 5 fractions) delivered during week 2. Tumour hypoxic volume (HV) was measured using 18F-fluoromisonidazole positron-emission tomography-computed tomography at baseline and following 1 week of buparlisib. RESULTS: Twenty-one patients were recruited with 9 patients evaluable for maximum tolerated dose (MTD) analysis. No dose-limiting toxicity was reported; therefore, 100 mg was declared the MTD, and 10 patients received this dose in the expansion phase. Ninety-four percent of treatment-related adverse events were ≤grade 2 with fatigue (67%), nausea (24%) and decreased appetite (19%) most common per patient. One serious adverse event (grade 3 hypoalbuminaemia) was possibly related to buparlisib. No unexpected radiotherapy toxicity was reported. Ten (67%) of 15 patients evaluable for imaging analysis were responders with 20% median reduction in HV at the MTD. CONCLUSION: This is the first clinical trial to combine a PI3K inhibitor with radiotherapy in NSCLC and investigate the effects of PI3K inhibition on tumour hypoxia. This combination was well tolerated and PI3K inhibition reduced hypoxia, warranting investigation into whether this novel class of radiosensitisers can improve radiotherapy outcomes.


Subject(s)
Adenocarcinoma of Lung/therapy , Aminopyridines/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , Lung Neoplasms/therapy , Morpholines/therapeutic use , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Tumor Hypoxia , Adenocarcinoma of Lung/diagnostic imaging , Adenocarcinoma of Lung/metabolism , Aged , Anorexia/chemically induced , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Chemoradiotherapy , Fatigue/chemically induced , Female , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Misonidazole/analogs & derivatives , Nausea/chemically induced , Positron Emission Tomography Computed Tomography , Radiotherapy
11.
Cell Death Dis ; 9(11): 1089, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356039

ABSTRACT

'Targeted' or 'biological' cancer treatments rely on differential gene expression between normal tissue and cancer, and genetic changes that render tumour cells especially sensitive to the agent being applied. Problems exist with the application of many agents as a result of damage to local tissues, tumour evolution and treatment resistance, or through systemic toxicity. Hence, there is a therapeutic need to uncover specific clinical targets which enhance the efficacy of cancer treatment whilst minimising the risk to healthy tissues. T-LAK cell-originated protein kinase (TOPK) is a MAPKK-like kinase which plays a role in cell cycle regulation and mitotic progression. As a consequence, TOPK expression is minimal in differentiated cells, although its overexpression is a pathophysiological feature of many tumours. Hence, TOPK has garnered interest as a cancer-specific biomarker and biochemical target with the potential to enhance cancer therapy whilst causing minimal harm to normal tissues. Small molecule inhibitors of TOPK have produced encouraging results as a stand-alone treatment in vitro and in vivo, and are expected to advance into clinical trials in the near future. In this review, we present the current literature pertaining to TOPK as a potential clinical target and describe the progress made in uncovering its role in tumour development. Firstly, we describe the functional role of TOPK as a pro-oncogenic kinase, followed by a discussion of its potential as a target for the treatment of cancers with high-TOPK expression. Next, we provide an overview of the current preclinical progress in TOPK inhibitor discovery and development, with respect to future adaptation for clinical use.


Subject(s)
Indolizines/therapeutic use , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Quinolones/therapeutic use , Quinoxalines/therapeutic use , Thiophenes/therapeutic use , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Discovery/methods , Humans , Indolizines/pharmacology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Quinolones/pharmacology , Quinoxalines/pharmacology , Thiophenes/pharmacology , Treatment Outcome
12.
Cell Cycle ; 17(12): 1513-1523, 2018.
Article in English | MEDLINE | ID: mdl-30045664

ABSTRACT

Cyclin-dependent kinase 1 (CDK1) orchestrates the transition from the G2 phase into mitosis and as cancer cells often display enhanced CDK1 activity, it has been proposed as a tumor specific anti-cancer target. Here we show that the effects of CDK1 inhibition are not restricted to tumor cells but can also reduce viability in non-cancer cells and sensitize them to radiation in a cell cycle dependent manner. Radiosensitization by the specific CDK1 inhibitor, RO-3306, was determined by colony formation assays in three tumor lines (HeLa, T24, SQ20B) and three non-cancer lines (HFL1, MRC-5, RPE). Initial results showed that CDK1 inhibition radiosensitized tumor cells, but did not sensitize normal fibroblasts and epithelial cells in colony formation assays despite effective inhibition of CDK1 signaling. Further investigation showed that normal cells were less sensitive to CDK1 inhibition because they remained predominantly in G1 for a prolonged period when plated in colony formation assays. In contrast, inhibiting CDK1 a day after plating, when the cells were going through G2/M phase, reduced their clonogenic survival both with and without radiation. Our finding that inhibition of CDK1 can damage normal cells in a cell cycle dependent manner indicates that targeting CDK1 in cancer patients may lead to toxicity in normal proliferating cells. Furthermore, our finding that cell cycle progression becomes easily stalled in non-cancer cells under normal culture conditions has general implications for testing anti-cancer agents in these cells.


Subject(s)
CDC2 Protein Kinase/antagonists & inhibitors , Cell Division/drug effects , DNA Damage/drug effects , G2 Phase/drug effects , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Humans , Signal Transduction/drug effects
13.
Nucleic Acids Res ; 46(15): 7731-7746, 2018 09 06.
Article in English | MEDLINE | ID: mdl-29986057

ABSTRACT

The nuclear pore complex (NPC) machinery is emerging as an important determinant in the maintenance of genome integrity and sensitivity to DNA double-strand break (DSB)-inducing agents, such as ionising radiation (IR). In this study, using a high-throughput siRNA screen, we identified the central channel NPC protein Nup54, and concomitantly its molecular partners Nup62 and Nup58, as novel factors implicated in radiosensitivity. Nup54 depletion caused an increase in cell death by mitotic catastrophe after IR, and specifically enhanced both the duration of the G2 arrest and the radiosensitivity of cells that contained replicated DNA at the time of IR exposure. Nup54-depleted cells also exhibited increased formation of chromosome aberrations arisen from replicated DNA. Interestingly, we found that Nup54 is epistatic with the homologous recombination (HR) factor Rad51. Moreover, using specific DNA damage repair reporters, we observed a decreased HR repair activity upon Nup54 knockdown. In agreement with a role in HR repair, we also demonstrated a decreased formation of HR-linked DNA synthesis foci and sister chromatid exchanges after IR in cells depleted of Nup54. Our study reveals a novel role for Nup54 in the response to IR and the maintenance of HR-mediated genome integrity.


Subject(s)
DNA Replication , DNA/metabolism , Nuclear Pore Complex Proteins/metabolism , Recombinational DNA Repair , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , DNA/genetics , DNA Breaks, Double-Stranded/radiation effects , HeLa Cells , Humans , MCF-7 Cells , Nuclear Pore/genetics , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/genetics , RNA Interference , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation, Ionizing , Sister Chromatid Exchange/radiation effects
14.
Br J Cancer ; 117(4): 503-512, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28677687

ABSTRACT

BACKGROUND: Tumour-specific radiosensitising treatments may enhance the efficacy of radiotherapy without exacerbating side effects. In this study we determined the radiation response following depletion or inhibition of TOPK, a mitogen-activated protein kinase kinase family Ser/Thr protein kinase that is upregulated in many cancers. METHODS: Radiation response was studied in a wide range of cancer cell lines and normal cells using colony formation assays. The effect on cell cycle progression was assessed and the relationship between TOPK expression and therapeutic efficacy was studied in a cohort of 128 prostate cancer patients treated with radical radiotherapy. RESULTS: TOPK knockdown did not alter radiation response in normal tissues, but significantly enhanced radiosensitivity in cancer cells. This result was recapitulated in TOPK knockout cells and with the TOPK inhibitor, OTS964. TOPK depletion altered the G1/S transition and G2/M arrest in response to radiation. Furthermore, TOPK depletion increased chromosomal aberrations, multinucleation and apoptotic cell death after irradiation. These results suggest a possible role for TOPK in the radiation-induced DNA damage checkpoints. These findings have clinical relevance, as elevated TOPK protein expression was associated with poorer clinical outcomes in prostate cancer patients treated with radical radiotherapy. CONCLUSIONS: This study demonstrates that TOPK disruption may cause tumour-specific radiosensitisation in multiple different tumour types.


Subject(s)
Cell Cycle Checkpoints , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Recurrence, Local/metabolism , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/radiation effects , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Quinolones/pharmacology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Survival Rate
15.
Cancer Biol Ther ; 18(6): 425-432, 2017 06 03.
Article in English | MEDLINE | ID: mdl-28494188

ABSTRACT

The identification of genetic determinants that underpin tumor radioresistance can help the development of targeted radiosensitizers or aid personalization of radiotherapy treatment. Here we identify signal recognition particle 72kDa (SRP72) as a novel gene involved in radioresistance. Knockdown of SRP72 resulted in significant radiosensitization of HeLa (cervical), PSN-1 (pancreatic), and T24 (bladder), BT-549 (breast) and MCF7 (breast) tumor lines as measured by colony formation assays. SRP72 depletion also resulted in the radiosensitization of normal lung fibroblast cell lines (HFL1 and MRC-5), demonstrating that the effect is not restricted to tumor cells. Increased radiosensitivity was not due to impaired DNA damage signaling or repair as assessed by γ-H2AX foci formation. Instead SRP72 depletion was associated with elevated levels of apoptosis after irradiation, as measured by caspase 3/7 activity, PARP-cleavage and Annexin-V staining, and with an induction of the unfolded protein response. Together, our results show that SRP72 is a novel gene involved in radioresistance.


Subject(s)
Radiation Tolerance , Signal Recognition Particle/genetics , Apoptosis , Cell Survival/radiation effects , Gene Knockdown Techniques , HeLa Cells , Humans , MCF-7 Cells , Protein Biosynthesis , Signal Recognition Particle/metabolism , Unfolded Protein Response
16.
Neurology ; 88(2): 131-142, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27974645

ABSTRACT

OBJECTIVE: To investigate mitophagy in 5 patients with severe dominantly inherited optic atrophy (DOA), caused by depletion of OPA1 (a protein that is essential for mitochondrial fusion), compared with healthy controls. METHODS: Patients with severe DOA (DOA plus) had peripheral neuropathy, cognitive regression, and epilepsy in addition to loss of vision. We quantified mitophagy in dermal fibroblasts, using 2 high throughput imaging systems, by visualizing colocalization of mitochondrial fragments with engulfing autophagosomes. RESULTS: Fibroblasts from 3 biallelic OPA1(-/-) patients with severe DOA had increased mitochondrial fragmentation and mitochondrial DNA (mtDNA)-depleted cells due to decreased levels of OPA1 protein. Similarly, in siRNA-treated control fibroblasts, profound OPA1 knockdown caused mitochondrial fragmentation, loss of mtDNA, impaired mitochondrial function, and mitochondrial mislocalization. Compared to controls, basal mitophagy (abundance of autophagosomes colocalizing with mitochondria) was increased in (1) biallelic patients, (2) monoallelic patients with DOA plus, and (3) OPA1 siRNA-treated control cultures. Mitophagic flux was also increased. Genetic knockdown of the mitophagy protein ATG7 confirmed this by eliminating differences between patient and control fibroblasts. CONCLUSIONS: We demonstrated increased mitophagy and excessive mitochondrial fragmentation in primary human cultures associated with DOA plus due to biallelic OPA1 mutations. We previously found that increased mitophagy (mitochondrial recycling) was associated with visual loss in another mitochondrial optic neuropathy, Leber hereditary optic neuropathy (LHON). Combined with our LHON findings, this implicates excessive mitochondrial fragmentation, dysregulated mitophagy, and impaired response to energetic stress in the pathogenesis of mitochondrial optic neuropathies, potentially linked with mitochondrial mislocalization and mtDNA depletion.


Subject(s)
GTP Phosphohydrolases/genetics , Mitophagy/genetics , Mutation/genetics , Optic Atrophy/genetics , Antioxidants/pharmacology , Cells, Cultured , Cognition Disorders/etiology , DNA Mutational Analysis , DNA, Mitochondrial/genetics , Family Health , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Male , Membrane Potential, Mitochondrial/genetics , Mitochondrial Proteins/genetics , Optic Atrophy/complications , Optic Atrophy/pathology , Pedigree , Protein Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquitin-Protein Ligases/genetics
17.
Oncotarget ; 6(8): 5978-89, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25788274

ABSTRACT

Colony formation is the gold standard assay for determining reproductive cell death after radiation treatment, since effects on proliferation often do not reflect survival. We have developed a high-throughput radiosensitivity screening method based on clonogenicity and screened a siRNA library against kinases. Thiamine pyrophosphokinase-1 (TPK1), a key component of Vitamin B1/thiamine metabolism, was identified as a target for radiosensitization. TPK1 knockdown caused significant radiosensitization in cancer but not normal tissue cell lines. Other means of blocking this pathway, knockdown of thiamine transporter-1 (THTR1) or treatment with the thiamine analogue pyrithiamine hydrobromide (PyrH) caused significant tumor specific radiosensitization. There was persistent DNA damage in cells irradiated after TPK1 and THTR1 knockdown or PyrH treatment. Thus this screen allowed the identification of thiamine metabolism as a novel radiosensitization target that affects DNA repair. Short-term modulation of thiamine metabolism could be a clinically exploitable strategy to achieve tumor specific radiosensitization.


Subject(s)
Neoplasms/metabolism , Neoplasms/radiotherapy , Thiamine/metabolism , Cell Line, Tumor , Colony-Forming Units Assay , DNA Damage , HCT116 Cells , HeLa Cells , High-Throughput Screening Assays , Humans , Pyrithiamine/pharmacology , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Thiamin Pyrophosphokinase/metabolism , Transfection
18.
Cancer Treat Rev ; 40(1): 109-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23583268

ABSTRACT

The ataxia telangiectasia and Rad3-related (ATR) plays an important role in maintaining genome integrity during DNA replication through the phosphorylation and activation of Chk1 and regulation of the DNA damage response. Preclinical studies have shown that disruption of ATR pathway can exacerbate the levels of replication stress in oncogene-driven murine tumors to promote cell killing. Additionally, inhibition of ATR can sensitise tumor cells to radiation or chemotherapy. Accumulating evidence suggests that targeting ATR can selectively sensitize cancer cells but not normal cells to DNA damage. Furthermore, in hypoxic conditions, ATR blockade results in overloading replication stress and DNA damage response causing cell death. Despite the attractiveness of ATR inhibition in the treatment of cancer, specific ATR inhibitors have remained elusive. In the last two years however, selective ATR inhibitors suitable for in vitro and - most recently - in vivo studies have been identified. In this article, we will review the literature on ATR function, its role in DDR and the potential of ATR inhibition to enhance the efficacy of radiation and chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Checkpoint Kinase 1 , Clinical Trials as Topic , Combined Modality Therapy , DNA Damage , DNA Replication , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Signal Transduction
19.
Cancer Biol Ther ; 13(11): 1072-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22825331

ABSTRACT

DNA damaging agents such as radiotherapy and gemcitabine are frequently used for the treatment of pancreatic cancer. However, these treatments typically provide only modest benefit. Improving the low survival rate for pancreatic cancer patients therefore remains a major challenge in oncology. Inhibition of the key DNA damage response kinase ATR has been suggested as an attractive approach for sensitization of tumor cells to DNA damaging agents, but specific ATR inhibitors have remained elusive. Here we investigated the sensitization potential of the first highly selective and potent ATR inhibitor, VE-821, in vitro. VE-821 inhibited radiation- and gemcitabine-induced phosphorylation of Chk1, confirming inhibition of ATR signaling. Consistently, VE-821 significantly enhanced the sensitivity of PSN-1, MiaPaCa-2 and primary PancM pancreatic cancer cells to radiation and gemcitabine under both normoxic and hypoxic conditions. ATR inhibition by VE-821 led to inhibition of radiation-induced G 2/M arrest in cancer cells. Reduced cancer cell radiosurvival following treatment with VE-821 was also accompanied by increased DNA damage and inhibition of homologous recombination repair, as evidenced by persistence of γH2AX and 53BP1 foci and inhibition of Rad51 foci, respectively. These findings support ATR inhibition as a novel approach to improve the efficacy and therapeutic index of standard cancer treatments across a large proportion of pancreatic cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Pyrazines/pharmacology , Sulfones/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Hypoxia/physiology , Cell Line, Tumor , Combined Modality Therapy , DNA Damage , DNA Repair , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazines/administration & dosage , Radiation-Sensitizing Agents/pharmacology , Signal Transduction , Sulfones/administration & dosage , Gemcitabine
20.
Carcinogenesis ; 33(11): 2026-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22822095

ABSTRACT

DNA polymerase eta (pol η) is the only DNA polymerase causally linked to carcinogenesis in humans. Inherited deficiency of pol η in the variant form of xeroderma pigmentosum (XPV) predisposes to UV-light-induced skin cancer. Pol η-deficient cells demonstrate increased sensitivity to cisplatin and oxaliplatin chemotherapy. We have found that XP30R0 fibroblasts derived from a patient with XPV are more resistant to cell kill by ionising radiation (IR) than the same cells complemented with wild-type pol η. This phenomenon has been confirmed in Burkitt's lymphoma cells, which either expressed wild-type pol η or harboured a pol η deletion. Pol η deficiency was associated with accumulation of cells in S-phase, which persisted after IR. Cells deficient in pol η demonstrated increased homologous recombination (HR)-directed repair of double strand breaks created by IR. Depletion of the HR protein, X-ray repair cross-complementing protein 3 (XRCC3), abrogated the radioresistance observed in pol η-deficient cells as compared with pol η-complemented cells. These findings suggest that HR mediates S-phase-dependent radioresistance associated with pol η deficiency. We propose that pol η protein levels in tumours may potentially be used to identify patients who require treatment with chemo-radiotherapy rather than radiotherapy alone for adequate tumour control.


Subject(s)
DNA-Directed DNA Polymerase/genetics , Homologous Recombination/genetics , Radiation Tolerance/genetics , S Phase/physiology , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Flow Cytometry , Gamma Rays , Humans , Neoplasms/metabolism , Neoplasms/pathology , Nucleic Acid Synthesis Inhibitors , RNA, Small Interfering/genetics , S Phase/radiation effects , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...