Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007759

ABSTRACT

The dominant role of non-homologous end-joining in the repair of radiation-induced double-strand breaks identifies DNA-dependent protein kinase (DNA-PK) as an excellent target for the development of radiosensitizers. We report the discovery of a new class of imidazo[4,5-c]pyridine-2-one DNA-PK inhibitors. Structure-activity studies culminated in the identification of 78 as a nM DNA-PK inhibitor with excellent selectivity for DNA-PK compared to related phosphoinositide 3-kinase (PI3K) and PI3K-like kinase (PIKK) families and the broader kinome, and displayed DNA-PK-dependent radiosensitization of HAP1 cells. Compound 78 demonstrated robust radiosensitization of a broad range of cancer cells in vitro, displayed high oral bioavailability, and sensitized colorectal carcinoma (HCT116/54C) and head and neck squamous cell carcinoma (UT-SCC-74B) tumor xenografts to radiation. Compound 78 also provided substantial tumor growth inhibition of HCT116/54C tumor xenografts in combination with radiation. Compound 78 represents a new, potent, and selective class of DNA-PK inhibitors with significant potential as radiosensitizers for cancer treatment.

2.
Br J Cancer ; 122(5): 603-612, 2020 03.
Article in English | MEDLINE | ID: mdl-31839676

ABSTRACT

The HER2-targeted antibody-drug conjugate trastuzumab emtansine (T-DM1) is approved for the treatment of metastatic, HER2-positive breast cancer after prior trastuzumab and taxane therapy, and has also demonstrated efficacy in the adjuvant setting in incomplete responders to neoadjuvant therapy. Despite its objective activity, intrinsic and acquired resistance to T-DM1 remains a major clinical challenge. T-DM1 mediates its activity in a number of ways, encompassing HER2 signalling blockade, Fc-mediated immune response and payload-mediated microtubule poisoning. Resistance mechanisms relating to each of these features have been demonstrated, and we outline the findings of these studies in this review. In our overview of the substantial literature on T-DM1 activity and resistance, we conclude that the T-DM1 resistance mechanisms most strongly supported by the experimental data relate to dysfunctional intracellular metabolism of the construct and subversion of DM1-mediated cell killing. Loss of dependence on signalling initiated by HER2-HER2 homodimers is not substantiated as a resistance mechanism by clinical or experimental studies, and the impact of EGFR expression and tumour immunological status requires further investigation. These findings are instructive with respect to strategies that might overcome T-DM1 resistance, including the use of second-generation anti-HER2 antibody-drug conjugates that deploy alternative linker-payload chemistries.


Subject(s)
Ado-Trastuzumab Emtansine/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Receptor, ErbB-2/biosynthesis , Ado-Trastuzumab Emtansine/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Clinical Trials, Phase III as Topic , Drug Resistance, Neoplasm , Female , Humans , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Randomized Controlled Trials as Topic , Receptor, ErbB-2/metabolism
3.
Biochem Pharmacol ; 169: 113641, 2019 11.
Article in English | MEDLINE | ID: mdl-31541630

ABSTRACT

DNA-dependent protein kinase (DNA-PK) plays a key role in repair of radiation-induced DNA double strand breaks (DSB) by non-homologous end-joining. DNA-PK inhibitors (DNA-PKi) are therefore efficient radiosensitisers, but normal tissue radiosensitisation represents a risk for their use in radiation oncology. Here we describe a novel prodrug, SN38023, that is metabolised to a potent DNA-PKi (IC87361) selectively in radioresistant hypoxic cells. DNA-PK inhibitory potency of SN38023 was 24-fold lower than IC87361 in cell-free assays, consistent with molecular modelling studies suggesting that SN38023 is unable to occupy one of the predicted DNA-PK binding modes of IC87361. One-electron reduction of the prodrug by radiolysis of anoxic formate solutions, and by metabolic reduction in anoxic HCT116/POR cells that overexpress cytochrome P450 oxidoreductase (POR), generated IC87361 efficiently as assessed by LC-MS. SN38023 inhibited radiation-induced Ser2056 autophosphorylation of DNA-PK catalytic subunit and radiosensitised HCT116/POR and UT-SCC-54C cells selectively under anoxia. SN38023 was an effective radiosensitiser in anoxic HCT116 spheroids, demonstrating potential for penetration into hypoxic tumour tissue, but in spheroid co-cultures of high-POR and POR-null cells it showed no evidence of bystander effects resulting from local diffusion of IC87361. Pharmacokinetics of IC87361 and SN38023 at maximum achievable doses in NIH-III mice demonstrated sub-optimal exposure of UT-SCC-54C tumour xenografts and did not provide significant tumour radiosensitisation. In conclusion, SN38023 has potential for exploiting hypoxia for selective delivery of a potent DNA-PKi to the most radioresistant subpopulation of cells in tumours. However, prodrugs providing improved systemic pharmacokinetics and that release DNA-PKi that elicit bystander effects are needed to maximise therapeutic utility.


Subject(s)
Benzopyrans/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Morpholines/pharmacology , Prodrugs/pharmacology , Radiation-Sensitizing Agents/pharmacology , Cell Hypoxia , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Phosphorylation , Protein Kinase Inhibitors/pharmacology
4.
Int J Radiat Biol ; 95(12): 1597-1612, 2019 12.
Article in English | MEDLINE | ID: mdl-31490091

ABSTRACT

Background and purpose: Poly(ADP-ribose)polymerase-1 (PARP1) and DNA-dependent protein kinase (DNA-PK) play key roles in the repair of radiation-induced DNA double strand breaks, but it is unclear which is the preferred therapeutic target in radiotherapy. Here we compare small molecule inhibitors of both as radiosensitizers of head and neck squamous cell carcinoma (HNSCC) cell lines.Methods: Two PARP1 inhibitors (olaparib, veliparib) and two DNA-PK inhibitors (KU57788, IC87361) were tested in 14 HNSCC cell lines and two non-tumorigenic lines (HEK-293 and WI-38/Va-13), with drug exposure for 6 or 24 h post-irradiation, using regrowth assays. For three lines (UT-SCC-54C, -74B, -76B), radiosensitization was also assessed by clonogenic assay under oxia and acute (6 h) anoxia, and for 54C cells under chronic hypoxia (0.2% O2 for 48 h). Relationships between sensitizer enhancement ratios (SER) and gene expression, assessed by RNA sequencing, were evaluated.Results: The inhibitors were minimally cytotoxic in the absence of radiation, with 74B and 54C cells the most sensitive to both olaparib and KU57788. Median SER values for each inhibitor at 1.1 µM were 1.12 (range 1.02-1.24) for olaparib, 1.08 (1.04-1.13) for veliparib, 1.35 (1.10-1.64) for IC87361 and 1.77 (1.41-2.38) for KU57788. The higher SER values for the DNA-PK inhibitors were observed with all cell lines (except HEK-293) and all concentrations tested and were confirmed by clonogenic assay. Radiosensitization by the DNA-PK inhibitors correlated with expression of SLFN11 mRNA. Radiosensitization by IC87361 and olaparib was significantly enhanced under acute anoxia and chronic hypoxia.Conclusions: The DNA-PK inhibitors KU57788 and IC87361 are more effective radiosensitizers than the PARP-1 inhibitors olaparib and veliparib at non-cytotoxic concentrations in HNSCC cell cultures and their activity is enhanced by SLFN11 and hypoxia.


Subject(s)
DNA-Activated Protein Kinase/antagonists & inhibitors , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Time Factors
5.
PLoS One ; 12(3): e0174381, 2017.
Article in English | MEDLINE | ID: mdl-28328949

ABSTRACT

CCAAT/enhancer-binding protein beta (C/EBPß) is a transcription factor controlling a broad range of genes essential for homeostasis, including genes related to immune functions, inflammation, metabolism and growth. Monocyte chemoattractant protein-1-induced protein 1 (MCPIP1) also called as Regnase-1 is an RNase and has been shown to decrease the stability of short-lived transcripts coding for inflammation-related proteins, including IL-1ß, IL-6, IL-2, IL-8, IL-12b, IER-3, c-Rel. We found previously that the half-life of the C/EBPß transcript is regulated by MCPIP. To understand the mechanism driving down-regulation of C/EBPß by MCPIP1, we applied an in vitro cleavage assay, followed by a luciferase-reporter assay and RNA immunoprecipitation (RIP). We demonstrated that MCPIP1 recognizes regions of the 3'UTR of C/EBPß mRNA and promotes its decay by introducing direct endonucleolytic cleavage.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , RNA, Messenger/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Down-Regulation/genetics , Hep G2 Cells , Humans , Immunoprecipitation/methods , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-2/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-rel/metabolism
6.
Angiogenesis ; 20(3): 325-340, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28197812

ABSTRACT

Monocyte Chemoattractant protein-induced protein 1 (MCPIP1), also known as Regnase-1, is encoded by the ZC3H12a gene, and it mediates inflammatory processes by regulating the stability of transcripts coding for proinflammatory cytokines and controlling activity of transcription factors, such as NF-κB and AP1. We found that MCPIP1 transcript and protein levels are strongly downregulated in clear cell renal cell carcinoma (ccRCC) samples, which were derived from patients surgically treated for renal cancer compared to surrounded normal tissues. Using Caki-1 cells as a model, we analyzed the role of MCPIP1 in cancer development. We showed that MCPIP1 expression depends on the proteasome activity; however, hypoxia and hypoxia inducible factor 2 alfa (HIF2α) are key factors lowering MCPIP1 expression. Furthermore, we found that MCPIP1 negatively regulates HIF1α and HIF2α levels and in the case of the last one, the mechanism is based on the regulation of the half time of transcript coding for HIF2α. Enhanced expression of MCPIP1 in Caki-1 cells results in a downregulation of transcripts encoding VEGFA, GLUT1, and IL-6. Furthermore, MCPIP1 decreases the activity of mTOR and protein kinase B (Akt) in normoxic conditions. Taken together, MCPIP1 contributes to the ccRCC development.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Ribonucleases/metabolism , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Glucose Transporter Type 1/metabolism , Humans , Leupeptins/pharmacology , Male , Middle Aged , NF-kappa B/metabolism , Proteasome Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases/genetics , Signal Transduction/drug effects , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Invest Dermatol ; 136(8): 1599-1607, 2016 08.
Article in English | MEDLINE | ID: mdl-27180111

ABSTRACT

ZC3H12A, which encodes the RNase monocyte chemotactic protein-induced protein 1 (MCPIP1), is up-regulated in psoriatic skin and reduced to normal levels after clinical treatments with anti-IL-17A/IL-17R neutralizing antibodies. In IL-17A-stimulated keratinocytes, MCPIP1 is rapidly increased at the transcript and protein levels. Also, IL-17A was found to be the main inducer of ZC3H12A expression in keratinocytes treated with supernatants derived from a Streptococcus pyogenes-activated psoriatic ex vivo model based on the co-culture of psoriatic cutaneous lymphocyte-associated antigen (CLA(+)) T cells and lesional epidermal cells. Moreover, MCPIP1 was aberrantly distributed in the suprabasal layers of psoriatic epidermis. In psoriatic samples, IL-17A-stimulated epidermal cell suspensions showed an increased MCPIP1 expression, especially in the mid-differentiated cellular compartment. The knockdown of ZC3H12A showed that this RNase participates in the regulation of the mRNAs present in suprabasal differentiated keratinocytes. Furthermore, JAK/STAT3 inhibition prevented the IL-17A-dependent induction of MCPIP1. In the mouse model of imiquimod-induced psoriasis, Zc3h12a expression was abrogated in Il17ra(-/-) mice. These results support the notion that IL-17A-mediated induction of MCPIP1 is involved in the regulation of local altered gene expression in suprabasal epidermal layers in psoriasis.


Subject(s)
Epidermis/enzymology , Interleukin-17/pharmacology , Psoriasis/enzymology , Psoriasis/genetics , Ribonucleases/metabolism , Transcription Factors/metabolism , Aminoquinolines/chemistry , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Biopsy , Coculture Techniques , Epidermis/metabolism , Gene Silencing , Humans , Imiquimod , Inflammation , Keratinocytes/cytology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Psoriasis/drug therapy , Receptors, Interleukin-17/metabolism , Ribonucleases/genetics , STAT3 Transcription Factor/metabolism , Skin/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
PLoS One ; 10(7): e0133746, 2015.
Article in English | MEDLINE | ID: mdl-26214508

ABSTRACT

The current evidence suggests that beneficial effects of mesenchymal stem cells (MSCs) toward myocardial repair are largely due to paracrine actions of several factors. Although Monocyte chemoattractant protein-induced protein 1 (MCPIP1) is involved in the regulation of inflammatory response, apoptosis and angiogenesis, whether MCPIP1 plays any role in stem cell-induced cardiac repair has never been examined. By employing retroviral (RV)-transduced overexpression of MCPIP1, we investigated the impact of MCPIP1 on viability, apoptosis, proliferation, metabolic activity, proteome, secretome and differentiation capacity of murine bone marrow (BM) - derived MSCs. MCPIP1 overexpression enhanced angiogenic and cardiac differentiation of MSCs compared with controls as indicated by elevated expression of genes accompanying angiogenesis and cardiomyogenesis in vitro. The proangiogenic activity of MCPIP1-overexpressing MSCs (MCPIP1-MSCs) was also confirmed by increased capillary-like structure formation under several culture conditions. This increase in differentiation capacity was associated with decreased proliferation of MCPIP1-MSCs when compared with controls. MCPIP1-MSCs also expressed increased levels of proteins involved in angiogenesis, autophagy, and induction of differentiation, but not adverse inflammatory agents. We conclude that MCPIP1 enhances endothelial and cardiac differentiation of MSCs. Thus, modulating MCPIP1 expression may be a novel approach useful for enhancing the immune-regulatory, anti-apoptotic, anti-inflammatory and regenerative capacity of BM-derived MSCs for myocardial repair and regeneration of ischemic tissues.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/physiology , Ribonucleases/biosynthesis , Animals , Bone Marrow Cells/cytology , Cell Proliferation/physiology , Cells, Cultured , Mesenchymal Stem Cells/cytology , Mice , Myocytes, Cardiac/cytology , Ribonucleases/genetics
9.
Biochim Biophys Acta ; 1843(4): 780-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24418043

ABSTRACT

Monocyte chemoattractant protein-induced protein 1 (MCPIP1) encoded by the ZC3H12a gene (also known as Regnase-1) is involved in the regulation of degradation of mRNA of inflammatory modulators and for processing of pre-miRNA. These functions depend on the presence of the PIN domain. Moreover, MCPIP1 was described as a negative regulator of NF-κB and AP-1 signaling pathways although mechanisms underlying such activity remain unknown. We aimed at determining the role of MCPIP1 in adipogenesis. Here, we present evidence that Mcpip1 transcription is transiently activated during 3T3-L1 transition from pre- to adipocytes. However Mcpip1 protein expression is also strongly decreased at day one after induction of adipogenesis. Knockdown of Mcpip1 results in an upregulation of C/EBPß and PPARγ mRNAs, whereas overexpression of MCPIP1 reduces the level of both transcription factors and impairs adipogenesis. MCPIP1-dependend modulation of C/EBPß and PPARγ levels results in a modulation of the expression of downstream controlled genes. In addition, decreased C/EBPß, but not PPARγ, depends on the activity of the MCPIP1 PIN domain, which is responsible for RNase properties of this protein. Together, these data confirm that MCPIP1 is a key regulator of adipogenesis.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Ribonucleases/genetics , Transcription, Genetic , 3T3-L1 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cell Differentiation , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Mice , PPAR gamma/biosynthesis , Signal Transduction
10.
Biol Chem ; 394(1): 113-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23091270

ABSTRACT

Silver nanoparticles (AgNPs) have many biological applications in biomedicine, biotechnology and other life sciences. Depending on the size, shape and the type of carrier, AgNPs demonstrate different physical and chemical properties. AgNPs have strong antimicrobial, antiviral and antifungal activity, thus they are used extensively in a range of medical settings, particularly in wound dressings but also in cosmetics. This study was undertaken to examine the potential toxic effects of 15 nm polyvinylpyrrolidone-coated AgNPs on primary normal human epidermal keratinocytes (NHEK). Cells were treated with different concentrations of AgNPs and then cell viability, metabolic activity and other biological and biochemical aspects of keratinocytes functioning were studied. We observed that AgNPs decrease keratinocyte viability, metabolism and also proliferatory and migratory potential of these cells. Moreover, longer exposure resulted in activation of caspase 3/7 and DNA damage. Our studies show for the first time, that AgNPs may present possible danger for primary keratinocytes, concerning activation of genotoxic and cytotoxic processes depending on the concentration.


Subject(s)
Keratinocytes/drug effects , Metal Nanoparticles/chemistry , Silver/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Survival/drug effects , Cells, Cultured , DNA Damage , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Keratinocytes/metabolism , Silver/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...