Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Int J Mol Sci ; 25(3)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38338943

ABSTRACT

An apical component of the cell cycle checkpoint and DNA damage repair response is the ataxia-telangiectasia mutated (ATM) Ser/Thr protein kinase. A variant of ATM, Ser49Cys (rs1800054; minor allele frequency = 0.011), has been associated with an elevated risk of melanoma development; however, the functional consequence of this variant is not defined. ATM-dependent signalling in response to DNA damage has been assessed in a panel of patient-derived lymphoblastoid lines and primary human melanocytic cell strains heterozygous for the ATM Ser49Cys variant allele. The ATM Ser49Cys allele appears functional for acute p53-dependent signalling in response to DNA damage. Expression of the variant allele did reduce the efficacy of oncogene expression in inducing senescence. These findings demonstrate that the ATM 146C>G Ser49Cys allele has little discernible effect on the acute response to DNA damage but has reduced function observed in the chronic response to oncogene over-expression. Analysis of melanoma, naevus and skin colour genomics and GWAS analyses have demonstrated no association of this variant with any of these outcomes. The modest loss of function detected suggest that the variant may act as a modifier of other variants of ATM/p53-dependent signalling.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Melanoma , Humans , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Melanoma/genetics , Oncogenes , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
2.
Br J Cancer ; 130(7): 1196-1205, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38287178

ABSTRACT

BACKGROUND: 5-Fluorouracil (5-FU) remains a core component of systemic therapy for colorectal cancer (CRC). However, response rates remain low, and development of therapy resistance is a primary issue. Combinatorial strategies employing a second agent to augment the therapeutic effect of chemotherapy is predicted to reduce the incidence of treatment resistance and increase the durability of response to therapy. METHODS: Here, we employed quantitative proteomics approaches to identify novel druggable proteins and molecular pathways that are deregulated in response to 5-FU, which might serve as targets to improve sensitivity to chemotherapy. Drug combinations were evaluated using 2D and 3D CRC cell line models and an ex vivo culture model of a patient-derived tumour. RESULTS: Quantitative proteomics identified upregulation of the mitosis-associated protein Aurora B (AURKB), within a network of upregulated proteins, in response to a 24 h 5-FU treatment. In CRC cell lines, AURKB inhibition with the dihydrogen phosphate prodrug AZD1152, markedly improved the potency of 5-FU in 2D and 3D in vitro CRC models. Sequential treatment with 5-FU then AZD1152 also enhanced the response of a patient-derived CRC cells to 5-FU in ex vivo cultures. CONCLUSIONS: AURKB inhibition may be a rational approach to augment the effectiveness of 5-FU chemotherapy in CRC.


Subject(s)
Colorectal Neoplasms , Fluorouracil , Organophosphates , Quinazolines , Humans , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Apoptosis , Aurora Kinase B/pharmacology , Aurora Kinase B/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm
3.
Pigment Cell Melanoma Res ; 37(1): 45-50, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37614154

ABSTRACT

Treatment of melanomas with targeted and immunotherapies has proven effective, but resistance to both treatments is a common outcome leaving a high proportion of patients without effective alternative treatment options. Replication stress is a common feature of melanomas, and this is effectively targeted using a combination of checkpoint kinase 1 (CHK1) inhibitor and low-dose hydroxyurea (LDHU). This combination also promotes inflammatory and anti-tumour immune responses in vivo. Melanoma cell lines resistant to BRAF inhibitor (BRAFi) or immune checkpoint inhibitors (ICI) retain their sensitivity to CHK1i + LDHU, with sensitivity similar to that of parental tumours. In vivo, BRAFi-resistant and BRAFi-sensitive parental tumours produce an identical immune response with treatment.


Subject(s)
Melanoma , Humans , Melanoma/pathology , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Proto-Oncogene Proteins B-raf , Checkpoint Kinase 1/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor
4.
Cancers (Basel) ; 14(24)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36551637

ABSTRACT

Immune checkpoint blockade (ICB) is now standard of care for several metastatic epithelial cancers and prolongs life expectancy for a significant fraction of patients. A hostile tumor microenvironment (TME) induced by intrinsic oncogenic signaling induces an immunosuppressive niche that protects the tumor cells, limiting the durability and efficacy of ICB therapies. Addition of receptor tyrosine kinase inhibitors (RTKi) as potential modulators of an unfavorable local immune environment has resulted in moderate life expectancy improvement. Though the combination strategy of ICB and RTKi has shown significantly better results compared to individual treatment, the benefits and adverse events are additive whereas synergy of benefit would be preferable. There is therefore a need to investigate the potential of inhibitors other than RTKs to reduce malignant cell survival while enhancing anti-tumor immunity. In the last five years, preclinical studies have focused on using small molecule inhibitors targeting cell cycle and DNA damage regulators such as CDK4/6, CHK1 and poly ADP ribosyl polymerase (PARP) to selectively kill tumor cells and enhance cytotoxic immune responses. This review provides a comprehensive overview of the available drugs that attenuate immunosuppression and overcome hostile TME that could be used to boost FDA-approved ICB efficacy in the near future.

5.
Cancers (Basel) ; 13(15)2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34359633

ABSTRACT

Drugs selectively targeting replication stress have demonstrated significant preclinical activity, but this has not yet translated into an effective clinical treatment. Here we report that targeting increased replication stress with a combination of Checkpoint kinase 1 inhibitor (CHK1i) with a subclinical dose of hydroxyurea targets also promotes pro-inflammatory cytokine/chemokine expression that is independent of cGAS-STING pathway activation and immunogenic cell death in human and murine melanoma cells. In vivo, this drug combination induces tumour regression which is dependent on an adaptive immune response. It increases cytotoxic CD8+ T cell activity, but the major adaptive immune response is a pronounced NKT cell tumour infiltration. Treatment also promotes an immunosuppressive tumour microenvironment through CD4+ Treg and FoxP3+ NKT cells. The number of these accumulated during treatment, the increase in FoxP3+ NKT cells numbers correlates with the decrease in activated NKT cells, suggesting they are a consequence of the conversion of effector to suppressive NKT cells. Whereas tumour infiltrating CD8+ T cell PD-1 and tumour PD-L1 expression was increased with treatment, peripheral CD4+ and CD8+ T cells retained strong anti-tumour activity. Despite increased CD8+ T cell PD-1, combination with anti-PD-1 did not improve response, indicating that immunosuppression from Tregs and FoxP3+ NKT cells are major contributors to the immunosuppressive tumour microenvironment. This demonstrates that therapies targeting replication stress can be well tolerated, not adversely affect immune responses, and trigger an effective anti-tumour immune response.

6.
Oncogenesis ; 10(5): 41, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33993200

ABSTRACT

Defective DNA repair is being demonstrated to be a useful target in cancer treatment. Currently, defective repair is identified by specific gene mutations, however defective repair is a common feature of cancers without these mutations. DNA damage triggers cell cycle checkpoints that are responsible for co-ordinating cell cycle arrest and DNA repair. Defects in checkpoint signalling components such as ataxia telangiectasia mutated (ATM) occur in a low proportion of cancers and are responsible for reduced DNA repair and increased genomic instability. Here we have investigated the AURKA-PLK1 cell cycle checkpoint recovery pathway that is responsible for exit from the G2 phase cell cycle checkpoint arrest. We demonstrate that dysregulation of PP6 and AURKA maintained elevated PLK1 activation to promote premature exit from only ATM, and not ATR-dependent checkpoint arrest. Surprisingly, depletion of the B55α subunit of PP2A that negatively regulates PLK1 was capable of overcoming ATM and ATR checkpoint arrests. Dysregulation of the checkpoint recovery pathway reduced S/G2 phase DNA repair efficiency and increased genomic instability. We found a strong correlation between dysregulation of the PP6-AURKA-PLK1-B55α checkpoint recovery pathway with signatures of defective homologous recombination and increased chromosomal instability in several cancer types. This work has identified an unrealised source of G2 phase DNA repair defects and chromosomal instability that are likely to be sensitive to treatments targeting defective repair.

7.
Am J Cancer Res ; 10(10): 3406-3414, 2020.
Article in English | MEDLINE | ID: mdl-33163279

ABSTRACT

Human papillomavirus (HPV) is the main causative agent in cervical cancers. Recurrent cervical cancer is refractory to currently available treatments. Clearly there is an urgent unmet need to investigate new therapeutic strategies for both the newly diagnosed and recurrent patient populations. We have previously shown that the presence of HPV oncogenes sensitizes cells to inhibition of aurora kinases (AURKs), which induces mitotic delay eventually leading to apoptotic cell death. In this study, we explored whether a dual approach of combining an AURK inhibitor, MLN8237 (Alisertib), with a range of Bcl-2 family anti-apoptotic protein inhibitors would accelerate cancer cell killing. Enhanced and rapid cervical cancer cell killing was observed when Alisertib was combined with inhibitors of either Bcl-2 (Venetoclax), Bcl-XL (A1331852) or Mcl-1 (A1210477) proteins, likely by accelerating apoptosis during mitotic delay due to the loss of functional Bcl-2, Mcl-1, or Bcl-XL. This study presents a promising approach to treating aggressive cervical cancers and may apply to other HPV-related cancers.

8.
Mol Oncol ; 14(1): 22-41, 2020 01.
Article in English | MEDLINE | ID: mdl-31733171

ABSTRACT

Ultraviolet radiation-induced DNA mutations are a primary environmental driver of melanoma. The reason for this very high level of unrepaired DNA lesions leading to these mutations is still poorly understood. The primary DNA repair mechanism for UV-induced lesions, that is, the nucleotide excision repair pathway, appears intact in most melanomas. We have previously reported a postreplication repair mechanism that is commonly defective in melanoma cell lines. Here we have used a genome-wide approach to identify the components of this postreplication repair mechanism. We have used differential transcript polysome loading to identify transcripts that are associated with UV response, and then functionally assessed these to identify novel components of this repair and cell cycle checkpoint network. We have identified multiple interaction nodes, including global genomic nucleotide excision repair and homologous recombination repair, and previously unexpected MASTL pathway, as components of the response. Finally, we have used bioinformatics to assess the contribution of dysregulated expression of these pathways to the UV signature mutation load of a large melanoma cohort. We show that dysregulation of the pathway, especially the DNA damage repair components, are significant contributors to UV mutation load, and that dysregulation of the MASTL pathway appears to be a significant contributor to high UV signature mutation load.


Subject(s)
DNA Repair/radiation effects , DNA Replication/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/genetics , Melanoma/metabolism , Microtubule-Associated Proteins/metabolism , Polyribosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , DNA Replication/radiation effects , G2 Phase Cell Cycle Checkpoints/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Genome-Wide Association Study , Humans , Melanoma/genetics , Melanoma/pathology , Microtubule-Associated Proteins/genetics , Mutation , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Polyribosomes/genetics , Polyribosomes/radiation effects , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering , RNA-Seq , Recombinational DNA Repair , Ultraviolet Rays , Up-Regulation
10.
J Cell Sci ; 132(24)2019 12 13.
Article in English | MEDLINE | ID: mdl-31767623

ABSTRACT

Melanocytic cell interactions are integral to skin homeostasis, and affect the outcome of multiple diseases, including cutaneous pigmentation disorders and melanoma. By using automated-microscopy and machine-learning-assisted morphology analysis of primary human melanocytes in co-culture, we performed combinatorial interrogation of melanocyte genotypic variants and functional assessment of lentivirus-introduced mutations. Keratinocyte-induced melanocyte dendricity, an indicator of melanocyte differentiation, was reduced in the melanocortin 1 receptor (MC1R) R/R variant strain and by NRAS.Q61K and BRAF.V600E expression, while expression of CDK4.R24C and RAC1.P29S had no detectable effect. Time-lapse tracking of melanocytes in co-culture revealed dynamic interaction phenotypes and hyper-motile cell states that indicated that, in addition to the known role in activating mitogenic signalling, MEK-pathway-activating mutations may also allow melanocytes to escape keratinocyte control and increase their invasive potential. Expanding this combinatorial platform will identify other therapeutic target mutations and melanocyte genetic variants, as well as increase understanding of skin cell interactions.


Subject(s)
Fibroblasts/cytology , Keratinocytes/cytology , Melanocytes/cytology , Cell Communication/physiology , Cell Line , Cells, Cultured , Coculture Techniques , Female , Fibroblasts/physiology , Humans , Machine Learning , Signal Transduction/physiology
11.
Cancers (Basel) ; 11(9)2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31500184

ABSTRACT

The poor selectivity of standard cytotoxic chemotherapy regimens causes severe side-effects in patients and reduces the quality of life during treatment. Targeting cancer-specific vulnerabilities can improve response rates, increase overall survival and limit toxic side effects in patients. Oncogene-induced replication stress serves as a tumour specific vulnerability and rationale for the clinical development of inhibitors targeting the DNA damage response (DDR) kinases (CHK1, ATR, ATM and WEE1). CHK1 inhibitors (CHK1i) have served as the pilot compounds in this class and their efficacy in clinical trials as single agents has been disappointing. Initial attempts to combine CHK1i with chemotherapies agents that enhance replication stress (such as gemcitabine) were reported to be excessively toxic. More recently, it has emerged that combining CHK1i with subclinical doses of replication stress inducers is more effective, better tolerated and more compatible with immunotherapies. Here we focus on the lessons learned during the clinical development of CHK1i with the goal of improving the design of future clinical trials utilizing DDR inhibitors to target replication stress in cancer.

12.
Mol Oncol ; 13(7): 1503-1518, 2019 07.
Article in English | MEDLINE | ID: mdl-31044505

ABSTRACT

Drugs such as gemcitabine that increase replication stress are effective chemotherapeutics in a range of cancer settings. These drugs effectively block replication and promote DNA damage, triggering a cell cycle checkpoint response through the ATR-CHK1 pathway. Inhibiting this signalling pathway sensitises cells to killing by replication stress-inducing drugs. Here, we investigated the effect of low-level replication stress induced by low concentrations (> 0.2 mm) of the reversible ribonucleotide reductase inhibitor hydroxyurea (HU), which slows S-phase progression but has little effect on cell viability or proliferation. We demonstrate that HU effectively synergises with CHK1, but not ATR inhibition, in > 70% of melanoma and non-small-cell lung cancer cells assessed, resulting in apoptosis and complete loss of proliferative potential in vitro and in vivo. Normal fibroblasts and haemopoietic cells retain viability and proliferative potential following exposure to CHK1 inhibitor plus low doses of HU, but normal cells exposed to CHK1 inhibitor combined with submicromolar concentrations of gemcitabine exhibited complete loss of proliferative potential. The effects of gemcitabine on normal tissue correlate with irreversible ATR-CHK1 pathway activation, whereas low doses of HU reversibly activate CHK1 independently of ATR. The combined use of CHK1 inhibitor and subclinical HU also triggered an inflammatory response involving the recruitment of macrophages in vivo. These data indicate that combining CHK1 inhibitor with subclinical HU is superior to combination with gemcitabine, as it provides equal anticancer efficacy but with reduced normal tissue toxicity. These data suggest a significant proportion of melanoma and lung cancer patients could benefit from treatment with this drug combination.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Hydroxyurea/pharmacology , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/adverse effects , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Progression , Female , Humans , Hydroxyurea/adverse effects , Hydroxyurea/therapeutic use , Lung Neoplasms/pathology , Melanoma/pathology , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Gemcitabine
14.
Oral Oncol ; 86: 105-112, 2018 11.
Article in English | MEDLINE | ID: mdl-30409290

ABSTRACT

OBJECTIVES: Human papilloma virus (HPV) is the main culprit in cancers of the cervix, penis, anus, skin, eye and head and neck. Current treatments for HPV cancers have not altered survival outcomes for 30 years and there is a significant lack of targeted therapeutic agents in the management of advanced HPV-related HNSCC. Here we show that survival and maintenance of HPV-positive HNC cells relies on the continuous expression of the major HPV oncogene, E7, and that Aurora kinases are critical for survival of high-risk HPV-positive HNC cells. MATERIALS AND METHODS: To assess the role of HPV E7 on HNC cell survival, RNA interference (RNAi) of the E7 gene was initially performed. Using an Aurora kinase inhibitor, Alisertib, the role of Aurora kinases in the carcinogenesis of HPV E7 positive HNC tumour lines was then investigated. An in vivo HNC xenograft model was also utilised to assess loss of tumour volume in response to RNAi E7 gene silencing and Alisertib treatment. RESULTS: RNAi silencing of the HPV E7 gene inhibited the growth of HPV-positive HNC cells and in vivo tumour load. We show that HPV E7 oncogene expression confers sensitivity to Alisertib on HNC cells where Alisertib-mediated loss in in vitro cell viability and in vivo tumour load is dependent on E7 expression. Moreover, Aurora kinase inhibition induced degradation of MCL-1 in HPV E7-expressing HNC cells. CONCLUSION: Overall, we show that Aurora kinases are a novel therapeutic target for HPV-positive HNCs. It might be feasible to combine Aurora kinase and MCL-1 inhibitors for future HNC therapies.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/metabolism , Azepines/pharmacology , Azepines/therapeutic use , Female , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Human papillomavirus 16/genetics , Human papillomavirus 16/isolation & purification , Human papillomavirus 16/metabolism , Humans , Leupeptins/pharmacology , Leupeptins/therapeutic use , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Proteolysis/drug effects , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Interference , Xenograft Model Antitumor Assays
15.
EMBO Mol Med ; 10(9)2018 09.
Article in English | MEDLINE | ID: mdl-30108112

ABSTRACT

The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in in vivo models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers.


Subject(s)
Aneuploidy , Cell Cycle Proteins/metabolism , Cytokinesis , Mitosis , Nuclear Proteins/metabolism , Breast Neoplasms/pathology , CDC2 Protein Kinase/metabolism , Caspases/metabolism , Cell Cycle Proteins/genetics , Cell Death , Cell Line, Tumor , Cyclin B/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Models, Biological , Nuclear Proteins/genetics
16.
Oncotarget ; 9(9): 8206-8222, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29492189

ABSTRACT

Besides somatic mutations or drug efflux, epigenetic reprogramming can lead to acquired drug resistance. We recently have identified early stress-induced multi-drug tolerant cancer cells termed induced drug-tolerant cells (IDTCs). Here, IDTCs were generated using different types of cancer cell lines; melanoma, lung, breast and colon cancer. A common loss of the H3K4me3 and H3K27me3 and gain of H3K9me3 mark was observed as a significant response to drug exposure or nutrient starvation in IDTCs. These epigenetic changes were reversible upon drug holidays. Microarray, qRT-PCR and protein expression data confirmed the up-regulation of histone methyltransferases (SETDB1 and SETDB2) which contribute to the accumulation of H3K9me3 concomitantly in the different cancer types. Genome-wide studies suggest that transcriptional repression of genes is due to concordant loss of H3K4me3 and regional increment of H3K9me3. Conversely, genome-wide CpG site-specific DNA methylation showed no common changes at the IDTC state. This suggests that distinct histone methylation patterns rather than DNA methylation are driving the transition from parental to IDTCs. In addition, silencing of SETDB1/2 reversed multi drug tolerance. Alterations of histone marks in early multi-drug tolerance with an increment in H3K9me3 and loss of H3K4me3/H3K27me3 is neither exclusive for any particular stress response nor cancer type specific but rather a generic response.

17.
Sci Rep ; 8(1): 5144, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572477

ABSTRACT

Successive rounds of chemical modification in three generations of benzopyran molecules have shown to select for different mechanisms of actions and progressive increases in anti-cancer activity. In this study, we investigated the mechanism of action of the third-generation benzopyran compounds, TRX-E-002-1 and TRX-E-009-1. High-content screening of a panel of 240 cancer cell lines treated with TRX-E-009-1 demonstrated it has broad anti-cancer potential. Within this screen, melanoma cell lines showed a range of sensitivities and subsequently a second independent panel of 21 melanoma 3D spheroid lines were assessed for their responses to both TRX-E-002-1 and TRX-E-009-1 compounds. Time-lapse microscopy illustrated both of these compounds caused mitotic delays in treated cells, resulting in either mitotic slippage or apoptosis. This finding along with immunostaining, in vitro polymerization assays, and animal experiments in both athymic and immunocompetent mice, demonstrates that these third-generation benzopyran compounds are potent tubulin polymerization inhibitors in vitro and in vivo, and this is the molecular basis of their anti-cancer activity in melanoma. These findings indicate these BP compounds may offer a novel anti-microtubule strategy for cancer intervention and provides the basis for further investigation into biomarkers of clinical sensitivity.


Subject(s)
Benzopyrans , Flavonoids , Melanoma, Experimental/drug therapy , Tubulin Modulators , Animals , Benzopyrans/chemistry , Benzopyrans/pharmacology , Cell Line, Tumor , Flavonoids/chemistry , Flavonoids/pharmacology , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 24(12): 2901-2912, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29535131

ABSTRACT

Purpose: Checkpoint kinase 1 inhibitors (CHEK1i) have single-agent activity in vitro and in vivo Here, we have investigated the molecular basis of this activity.Experimental Design: We have assessed a panel of melanoma cell lines for their sensitivity to the CHEK1i GNE-323 and GDC-0575 in vitro and in vivo The effects of these compounds on responses to DNA replication stress were analyzed in the hypersensitive cell lines.Results: A subset of melanoma cell lines is hypersensitive to CHEK1i-induced cell death in vitro, and the drug effectively inhibits tumor growth in vivo In the hypersensitive cell lines, GNE-323 triggers cell death without cells entering mitosis. CHEK1i treatment triggers strong RPA2 hyperphosphorylation and increased DNA damage in only hypersensitive cells. The increased replication stress was associated with a defective S-phase cell-cycle checkpoint. The number and intensity of pRPA2 Ser4/8 foci in untreated tumors appeared to be a marker of elevated replication stress correlated with sensitivity to CHEK1i.Conclusions: CHEK1i have single-agent activity in a subset of melanomas with elevated endogenous replication stress. CHEK1i treatment strongly increased this replication stress and DNA damage, and this correlated with increased cell death. The level of endogenous replication is marked by the pRPA2Ser4/8 foci in the untreated tumors, and may be a useful marker of replication stress in vivoClin Cancer Res; 24(12); 2901-12. ©2018 AACR.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , DNA Replication/drug effects , Drug Resistance, Neoplasm , Melanoma/genetics , Melanoma/metabolism , Stress, Physiological , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Melanoma/drug therapy , Melanoma/pathology , Mice , Mitosis/drug effects , Mitosis/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Stress, Physiological/drug effects , Xenograft Model Antitumor Assays
19.
Cell Cycle ; 17(5): 652-668, 2018.
Article in English | MEDLINE | ID: mdl-28749250

ABSTRACT

We report for the first time the mechanism of action of the natural product thalicthuberine (TH) in prostate and cervical cancer cells. TH induced a strong accumulation of LNCaP cells in mitosis, severe mitotic spindle defects, and asymmetric cell divisions, ultimately leading to mitotic catastrophe accompanied by cell death through apoptosis. However, unlike microtubule-binding drugs (vinblastine and paclitaxel), TH did not directly inhibit tubulin polymerization when tested in a cell-free system, whereas it reduced cellular microtubule polymer mass in LNCaP cells. This suggests that TH indirectly targets microtubule dynamics through inhibition of a critical regulator or tubulin-associated protein. Furthermore, TH is not a major substrate for P-glycoprotein (Pgp), which is responsible for multidrug resistance in numerous cancers, providing a rationale to further study TH in cancers with Pgp-mediated treatment resistance. The identification of TH's molecular target in future studies will be of great value to the development of TH as potential treatment of multidrug-resistant tumors.


Subject(s)
Alkaloids/pharmacology , Antimitotic Agents/pharmacology , Apoptosis/drug effects , Phenanthrenes/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Microtubules/metabolism , Mitosis/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Spindle Apparatus/drug effects , Spindle Apparatus/physiology , Tubulin/genetics , Tubulin/metabolism
20.
Clin Cancer Res ; 24(5): 1090-1102, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29196297

ABSTRACT

Purpose: Identify and characterize novel combinations of sorafenib with anti-inflammatory painkillers to target difficult-to-treat RAS-mutant cancer.Experimental Design: The cytotoxicity of acetylsalicylic acid (aspirin) in combination with the multikinase inhibitor sorafenib (Nexavar) was assessed in RAS-mutant cell lines in vitro The underlying mechanism for the increased cytotoxicity was investigated using selective inhibitors and shRNA-mediated gene knockdown. In vitro results were confirmed in RAS-mutant xenograft mouse models in vivoResults: The addition of aspirin but not isobutylphenylpropanoic acid (ibruprofen) or celecoxib (Celebrex) significantly increased the in vitro cytotoxicity of sorafenib. Mechanistically, combined exposure resulted in increased BRAF/CRAF dimerization and the simultaneous hyperactivation of the AMPK and ERK pathways. Combining sorafenib with other AMPK activators, such as metformin or A769662, was not sufficient to decrease cell viability due to sole activation of the AMPK pathway. The cytotoxicity of sorafenib and aspirin was blocked by inhibition of the AMPK or ERK pathways through shRNA or via pharmacologic inhibitors of RAF (LY3009120), MEK (trametinib), or AMPK (compound C). The combination was found to be specific for RAS/RAF-mutant cells and had no significant effect in RAS/RAF-wild-type keratinocytes or melanoma cells. In vivo treatment of human xenografts in NSG mice with sorafenib and aspirin significantly reduced tumor volume compared with each single-agent treatment.Conclusions: Combination sorafenib and aspirin exerts cytotoxicity against RAS/RAF-mutant cells by simultaneously affecting two independent pathways and represents a promising novel strategy for the treatment of RAS-mutant cancers. Clin Cancer Res; 24(5); 1090-102. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aspirin/pharmacology , Neoplasms/drug therapy , Sorafenib/pharmacology , ras Proteins/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aspirin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mutation , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Sorafenib/therapeutic use , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...