Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biomedicines ; 9(7)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203351

ABSTRACT

Human epidermal growth factor receptor (EGFR) 2 (HER2) is overexpressed/amplified in about 25% of all breast cancers, and EGFR is overexpressed in up to 76% and amplified in up to 24% of triple-negative breast cancers (TNBC). Here, we aimed to identify inhibitors that may enhance the anti-tumor activity of neratinib for HER2+ breast cancer and TNBC. By conducting a non-biased high-throughput RNA interference screening, we identified PI3K/AKT/mTOR and MAPK as two potential inhibitory synergistic canonical pathways. We confirmed that everolimus (mTOR inhibitor) and trametinib (MEK inhibitor) enhances combinatorial anti-proliferative effects with neratinib under anchorage-independent growth conditions (p < 0.05). Compared to single agent neratinib, the combination therapies significantly enhanced tumor growth inhibition in both SUM190 HER2+ breast cancer (neratinib plus everolimus, 77%; neratinib plus trametinib, 77%; p < 0.0001) and SUM149 TNBC (neratinib plus everolimus, 71%; neratinib plus trametinib, 81%; p < 0.0001) xenograft models. Compared to single-agent neratinib, everolimus, or trametinib, both everolimus plus neratinib and trametinib plus neratinib significantly suppressed proliferation marker Ki67 and enhanced antitumor efficacy by activating the apoptosis pathway shown by increased Bim and cleaved-PARP expression. Taken together, our data justify new neratinib-based combinations for both HER2+ breast cancer and TNBC.

2.
NPJ Breast Cancer ; 7(1): 63, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34045483

ABSTRACT

Lapatinib (L) plus trastuzumab (T), with endocrine therapy for estrogen receptor (ER)+ tumors, but without chemotherapy, yielded meaningful response in HER2+ breast cancer (BC) neoadjuvant trials. The irreversible/pan-HER inhibitor neratinib (N) has proven more potent than L. However, the efficacy of N+T in comparison to pertuzumab (P) + T or L + T (without chemotherapy) remains less studied. To address this, mice bearing HER2+ BT474-AZ (ER+) cell and BCM-3963 patient-derived BC xenografts were randomized to vehicle, N, T, P, N+T, or P+T, with simultaneous estrogen deprivation for BT474-AZ. Time to tumor regression/progression and incidence/time to complete response (CR) were determined. Changes in key HER pathway and proliferative markers were assessed by immunohistochemistry and western blot of short-term-treated tumors. In the BT474-AZ model, while all N, P, T, N + T, and P + T treated tumors regressed, N + T-treated tumors regressed faster than P, T, and P + T. Further, N + T was superior to N and T alone in accelerating CR. In the BCM-3963 model, which was refractory to T, P, and P + T, while N and N + T yielded 100% CR, N + T accelerated the CR compared to N. Ki67, phosphorylated (p) AKT, pS6, and pERK levels were largely inhibited by N and N + T, but not by T, P, or P + T. Phosphorylated HER receptor levels were also markedly inhibited by N and N + T, but not by P + T or L + T. Our findings establish the efficacy of combining N with T and support clinical testing to investigate the efficacy of N + T with or without chemotherapy in the neoadjuvant setting for HER2+ BC.

3.
Br J Cancer ; 124(7): 1249-1259, 2021 03.
Article in English | MEDLINE | ID: mdl-33473169

ABSTRACT

BACKGROUND: Human epidermal growth factor 2 (HER2/ERBB2) is frequently amplified/mutated in cancer. The tyrosine kinase inhibitors (TKIs) lapatinib, neratinib, and tucatinib are FDA-approved for the treatment of HER2-positive breast cancer. Direct comparisons of the preclinical efficacy of the TKIs have been limited to small-scale studies. Novel biomarkers are required to define beneficial patient populations. METHODS: In this study, the anti-proliferative effects of the three TKIs were directly compared using a 115 cancer cell line panel. Novel TKI response/resistance markers were identified through cross-analysis of drug response profiles with mutation, gene copy number and expression data. RESULTS: All three TKIs were effective against HER2-amplified breast cancer models; neratinib showing the most potent activity, followed by tucatinib then lapatinib. Neratinib displayed the greatest activity in HER2-mutant and EGFR-mutant cells. High expression of HER2, VTCN1, CDK12, and RAC1 correlated with response to all three TKIs. DNA damage repair genes were associated with TKI resistance. BRCA2 mutations were correlated with neratinib and tucatinib response, and high expression of ATM, BRCA2, and BRCA1 were associated with neratinib resistance. CONCLUSIONS: Neratinib was the most effective HER2-targeted TKI against HER2-amplified, -mutant, and EGFR-mutant cell lines. This analysis revealed novel resistance mechanisms that may be exploited using combinatorial strategies.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Apoptosis , Cell Proliferation , Humans , Lapatinib/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Quinolines/pharmacology , Trastuzumab/pharmacology , Tumor Cells, Cultured
4.
Clin Cancer Res ; 27(6): 1681-1694, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33414137

ABSTRACT

PURPOSE: Neratinib is an irreversible, pan-HER tyrosine kinase inhibitor that is FDA approved for HER2-overexpressing/amplified (HER2+) breast cancer. In this preclinical study, we explored the efficacy of neratinib in combination with inhibitors of downstream signaling in HER2+ cancers in vitro and in vivo. EXPERIMENTAL DESIGN: Cell viability, colony formation assays, and Western blotting were used to determine the effect of neratinib in vitro. In vivo efficacy was assessed with patient-derived xenografts (PDX): two breast, two colorectal, and one esophageal cancer (with HER2 mutations). Four PDXs were derived from patients who received previous HER2-targeted therapy. Proteomics were assessed through reverse phase protein arrays and network-level adaptive responses were assessed through Target Score algorithm. RESULTS: In HER2+ breast cancer cells, neratinib was synergistic with multiple agents, including mTOR inhibitors everolimus and sapanisertib, MEK inhibitor trametinib, CDK4/6 inhibitor palbociclib, and PI3Kα inhibitor alpelisib. We tested efficacy of neratinib with everolimus, trametinib, or palbociclib in five HER2+ PDXs. Neratinib combined with everolimus or trametinib led to a 100% increase in median event-free survival (EFS; tumor doubling time) in 25% (1/4) and 60% (3/5) of models, respectively, while neratinib with palbociclib increased EFS in all five models. Network analysis of adaptive responses demonstrated upregulation of EGFR and HER2 signaling in response to CDK4/6, mTOR, and MEK inhibition, possibly providing an explanation for the observed synergies with neratinib. CONCLUSIONS: Taken together, our results provide strong preclinical evidence for combining neratinib with CDK4/6, mTOR, and MEK inhibitors for the treatment of HER2+ cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Breast Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Esophageal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Everolimus/administration & dosage , Female , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Piperazines/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Pyrimidinones/administration & dosage , Quinolines/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Nucleosides Nucleotides Nucleic Acids ; 39(1-3): 407-425, 2020.
Article in English | MEDLINE | ID: mdl-32310030

ABSTRACT

Telomerase activity has been regarded as a critical step in cellular immortalization and carcinogenesis and because of this, regulation of telomerase represents an attractive target for anti-tumor specific therapeutics. Recently, one avenue of cancer research focuses on antisense strategy to target the oncogenes or cancer driver genes, in a sequence specific fashion to down-regulate the expression of the target gene. The protein catalytic subunit, human telomerase reverse transcriptase (hTERT) and the template RNA component (hTERC) are essential for telomerase function, thus theoretically, inhibition of telomerase activity can be achieved by interfering with either the gene expression of hTERT or the hTERC of the telomerase enzymatic complex. The present study showed that phosphorothioate antisense oligonucleotide (sASO)-nuclear localization signal (NLS) peptide conjugates targeting hTERC could inhibit telomerase activity very efficiently at 5 µM concentration but less efficiently at 1 µM concentration. On the other hand, siRNA targeting hTERT mRNA could strongly suppress hTERT expression at 200 nM concentration. It was also revealed that siRNA targeting hTERT could induce telomere attrition and then irreversible arrest of proliferation of cancer cells.


Subject(s)
Cell Cycle Checkpoints/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nuclear Localization Signals/chemistry , Oligonucleotides, Antisense , Phosphates/chemistry , Telomerase/antagonists & inhibitors , Telomere/chemistry , Cell Proliferation/drug effects , Enzyme Activation , HeLa Cells , Humans , Peptides/chemistry , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Telomerase/chemistry , Tumor Cells, Cultured
6.
Cancer Discov ; 10(5): 674-687, 2020 05.
Article in English | MEDLINE | ID: mdl-32213539

ABSTRACT

Amplification of and oncogenic mutations in ERBB2, the gene encoding the HER2 receptor tyrosine kinase, promote receptor hyperactivation and tumor growth. Here we demonstrate that HER2 ubiquitination and internalization, rather than its overexpression, are key mechanisms underlying endocytosis and consequent efficacy of the anti-HER2 antibody-drug conjugates (ADC) ado-trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) in lung cancer cell lines and patient-derived xenograft models. These data translated into a 51% response rate in a clinical trial of T-DM1 in 49 patients with ERBB2-amplified or -mutant lung cancers. We show that cotreatment with irreversible pan-HER inhibitors enhances receptor ubiquitination and consequent ADC internalization and efficacy. We also demonstrate that ADC switching to T-DXd, which harbors a different cytotoxic payload, achieves durable responses in a patient with lung cancer and corresponding xenograft model developing resistance to T-DM1. Our findings may help guide future clinical trials and expand the field of ADC as cancer therapy. SIGNIFICANCE: T-DM1 is clinically effective in lung cancers with amplification of or mutations in ERBB2. This activity is enhanced by cotreatment with irreversible pan-HER inhibitors, or ADC switching to T-DXd. These results may help address unmet needs of patients with HER2-activated tumors and no approved targeted therapy.See related commentary by Rolfo and Russo, p. 643.This article is highlighted in the In This Issue feature, p. 627.


Subject(s)
Lung Neoplasms/drug therapy , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Mutation
7.
Clin Cancer Res ; 26(10): 2393-2403, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32034078

ABSTRACT

PURPOSE: Evaluating drug responses using primary patient-derived cells ex vivo represents a potentially rapid and efficient approach to screening for new treatment approaches. Here, we sought to identify neratinib combinations in HER2 mutant non-small cell lung cancer (NSCLC) patient xenograft-derived organotypic spheroids (XDOTS) using a short-term ex vivo system. EXPERIMENTAL DESIGN: We generated two HER2-mutant NSCLC PDX models [DFCI359 (HER2 exon19 755_757LREdelinsRP) and DFCI315 (HER2 exon20 V777_G778insGSP)] and used the PDX tumors to generate XDOTS. Tumor spheroids were grown in a microfluidic device and treated ex vivo with neratinib-based drug combinations. Live/dead quantification was performed by dual-labeling deconvolution fluorescence microscopy. The most efficacious ex vivo combination was subsequently validated in vivo using the DFCI359 and DFCI315 PDXs and a HER2 YVMA genetically engineered mouse model. RESULTS: Both neratinib and afatinib, but not gefitinib, induced cell death in DFCI359 XDOTS. The combinations of neratinib/trastuzumab and neratinib/temsirolimus enhanced the therapeutic benefit of neratinib alone in DFCI315 and DFCI359. The combination of neratinib and trastuzumab in vivo was more effective compared with single-agent neratinib or trastuzumab and was associated with more robust inhibition of HER2 and downstream signaling. CONCLUSIONS: The XDOTS platform can be used to evaluate therapies and therapeutic combinations ex vivo using PDX tumors. This approach may accelerate the identification and clinical development of therapies for targets with no or few existing models and/or therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mutation , Receptor, ErbB-2/genetics , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Quinolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Spheroids, Cellular , Trastuzumab/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Pharmacol Res Perspect ; 7(5): e00521, 2019 10.
Article in English | MEDLINE | ID: mdl-31523434

ABSTRACT

Diarrhea is one of the most commonly reported adverse effect of hemotherapy and targeted cancer therapies, such as tyrosine kinase inhibitors (TKI), which often significantly impact patient quality of life, morbidity, and mortality. Neratinib is an oral, irreversible pan-HER tyrosine kinase inhibitor, which is clinically active in HER2-positive breast cancer. Diarrhea is the most common side effect of this potent anticancer drug and the reasons for this adverse effect are still largely unclear. We have recently shown that activation of the calcium-sensing Receptor (CaSR) can inhibit secretagogue-induced diarrhea in the colon, therefore we hypothesized that CaSR activation may also mitigate neratinib-induced diarrhea. Using an established ex vivo model of isolated intestinal segments, we investigated neratinib-induced fluid secretion and the ability of CaSR activation to abate the secretion. In our study, individual segments of the rat intestine (proximal, middle, distal small intestine, and colon) were procured and perfused intraluminally with various concentrations of neratinib (10, 50, 100 nmol L-1). In a second set of comparison experiments, intraluminal calcium concentration was modulated (from 1.0 to 5.0 or 7.0 mmol L-1), both pre- and during neratinib exposure. In a separate series of experiments R-568, a known calcimimetic was used CaSR activation and effect was compared to elevated Ca2+ concentration (5.0 and 7.0 mmol L-1). As a result, CaSR activation with elevated Ca2+ concentration (5.0 and 7.0 mmol L-1) or R-568 markedly reduced neratinib-induced fluid secretion in a dose-dependent manner. Pre-exposure to elevated luminal calcium solutions (5.0 and 7.0 mmol L-1) also prevented neratinib-induced fluid secretion. In conclusion, exposure to luminal neratinib resulted in a pronounced elevation in fluid secretion in the rat intestine. Increasing luminal calcium inhibits the neratinib-associated fluid secretion in a dose-dependent manner. These results suggest that CaSR activation may be a potent therapeutic target to reduce chemotherapy-associated diarrhea.


Subject(s)
Diarrhea/drug therapy , Phenethylamines/pharmacology , Propylamines/pharmacology , Quinolines/adverse effects , Receptors, Calcium-Sensing/metabolism , Animals , Calcium/metabolism , Diarrhea/chemically induced , Diarrhea/metabolism , Diarrhea/prevention & control , Dose-Response Relationship, Drug , Intestines , Male , Perfusion , Rats , Up-Regulation
9.
Breast Cancer Res ; 21(1): 94, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409375

ABSTRACT

BACKGROUND: Human epidermal growth factor receptor-2 (HER2)-targeted therapies prolong survival in HER2-positive breast cancer patients. Benefit stems primarily from improved control of systemic disease, but up to 50% of patients progress to incurable brain metastases due to acquired resistance and/or limited permeability of inhibitors across the blood-brain barrier. Neratinib, a potent irreversible pan-tyrosine kinase inhibitor, prolongs disease-free survival in the extended adjuvant setting, and several trials evaluating its efficacy alone or combination with other inhibitors in early and advanced HER2-positive breast cancer patients are ongoing. However, its efficacy as a first-line therapy against HER2-positive breast cancer brain metastasis has not been fully explored, in part due to the lack of relevant pre-clinical models that faithfully recapitulate this disease. Here, we describe the development and characterisation of a novel syngeneic model of spontaneous HER2-positive breast cancer brain metastasis (TBCP-1) and its use to evaluate the efficacy and mechanism of action of neratinib. METHODS: TBCP-1 cells were derived from a spontaneous BALB/C mouse mammary tumour and characterised for hormone receptors and HER2 expression by flow cytometry, immunoblotting and immunohistochemistry. Neratinib was evaluated in vitro and in vivo in the metastatic and neoadjuvant setting. Its mechanism of action was examined by transcriptomic profiling, function inhibition assays and immunoblotting. RESULTS: TBCP-1 cells naturally express high levels of HER2 but lack expression of hormone receptors. TBCP-1 tumours maintain a HER2-positive phenotype in vivo and give rise to a high incidence of spontaneous and experimental metastases in the brain and other organs. Cell proliferation/viability in vitro is inhibited by neratinib and by other HER2 inhibitors, but not by anti-oestrogens, indicating phenotypic and functional similarities to human HER2-positive breast cancer. Mechanistically, neratinib promotes a non-apoptotic form of cell death termed ferroptosis. Importantly, metastasis assays demonstrate that neratinib potently inhibits tumour growth and metastasis, including to the brain, and prolongs survival, particularly when used as a neoadjuvant therapy. CONCLUSIONS: The TBCP-1 model recapitulates the spontaneous spread of HER2-positive breast cancer to the brain seen in patients and provides a unique tool to identify novel therapeutics and biomarkers. Neratinib-induced ferroptosis provides new opportunities for therapeutic intervention. Further evaluation of neratinib neoadjuvant therapy is warranted.


Subject(s)
Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Ferroptosis/drug effects , Quinolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Female , Gene Expression Profiling , Immunohistochemistry , Isografts , Mice , Molecular Targeted Therapy , Neoadjuvant Therapy , Quinolines/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
10.
J Cell Physiol ; 234(4): 4874-4887, 2019 04.
Article in English | MEDLINE | ID: mdl-30203445

ABSTRACT

Regorafenib is approved for the treatment of colorectal cancer and hepatocellular carcinoma. In the trial NCT02466802, we have discovered that regorafenib can be safely combined with the phosphodiesterase 5 inhibitor sildenafil in advanced solid tumor patients. The present studies determined whether the approved ERBB1/2/4 and RAS downregulating drug neratinib, could enhance the lethality of [regorafenib + sildenafil]. Neratinib enhanced [regorafenib + sildenafil] lethality in a greater than additive fashion in colon cancer cells. The drug combination reduced the expression of mutant K-RAS and of multiple histone deacetylase (HDAC) proteins that required autophagosome formation. It caused green fluorescent protein or red fluorescent protein-tagged forms of K-RAS V12 to localize into large intracellular vesicles. Compared with [regorafenib + sildenafil], the three-drug combination caused greater and more prolonged activation of the ATM-AMPK-ULK-1 pathway and caused a greater suppression and prolonged inactivation of mammalian target of rapamycin, AKT, and p70 S6K. Approximately 70% of enhanced lethality caused by neratinib required ataxia-telangiectasia-mutated (ATM)-AMP-dependent protein kinase (AMPK) signaling whereas knockdown of Beclin1, ATG5, FADD, and CD95 completely prevented the elevated killing effect. Exposure of cells to [regorafenib + sildenafil] reduced the expression of the checkpoint immunotherapy biomarkers programmed death-ligand 1, ornithine decarboxylase, and indoleamine 2,3-dioxygenase-1 and increased the expression of major histocompatibility complex A (MHCA), which also required autophagosome formation. Knockdown of specific HDAC proteins recapitulated the effects observed using chemical agents. In vivo, using mouse cancer models, neratinib significantly enhanced the antitumor efficacy of [regorafenib + sildenafil]. Our data support performing a new three drug Phase I trial combining regorafenib, sildenafil, and neratinib.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Sildenafil Citrate/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Synergism , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphodiesterase 5 Inhibitors/pharmacology , Xenograft Model Antitumor Assays
11.
Cancer Biol Ther ; 20(2): 157-168, 2019.
Article in English | MEDLINE | ID: mdl-30183517

ABSTRACT

Cancers expressing mutant RAS are associated with a weaker response to chemotherapy and a shorter overall patient survival. We have demonstrated that the irreversible inhibitor of ERBB1/2/4, neratinib, inhibits ERBB1/2/4 and causes their internalization and autolysosomal degradation. Fellow-traveler membrane proteins with RTKs, including mutant K-/N-RAS, were also degraded. We discovered that the CDK4/6 inhibitor palbociclib increased autophagosome and then autolysosome levels in a time dependent fashion, did not reduce mTOR activity, and interacted with temsirolimus to kill. Neratinib and palbociclib interacted in a greater than additive manner to increase autophagosome and then autolysosome levels in a time dependent fashion, and to cause tumor cell killing. Killing required the expression of ATM and AMPKα, Beclin1 and ATG5, BAX and BAK and of AIF, but not of caspase 9. In some cells over-expression of BCL-XL was protective whereas in others it was ineffective. The lethality of [neratinib + palbociclib] was modestly enhanced by the PDE5 inhibitor sildenafil and strongly enhanced by the HDAC inhibitor sodium valproate. This was associated with K-RAS degradation and a greater than additive increase in autophagosome and autolysosome levels. Killing by the three-drug combination required ATM and AMPKα, and, to a greater extent, Beclin1 and ATG5. In vivo, [valproate + palbociclib] and [neratinib + valproate + palbociclib] interacted to suppress the growth of a carboplatin/paclitaxel resistant PDX ovarian tumors that express a mutant N-RAS. Our data support performing a future three-drug trial with these agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Valproic Acid/pharmacology , Animals , Drug Synergism , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Ovarian Neoplasms/drug therapy , Piperazines/administration & dosage , Pyridines/administration & dosage , Quinolines/administration & dosage , Valproic Acid/administration & dosage , Xenograft Model Antitumor Assays
12.
Anal Sci ; 29(9): 927-30, 2013.
Article in English | MEDLINE | ID: mdl-24025579

ABSTRACT

The tumor necrosis factor-α (TNF-α) is implicated in periodontal disease, and there was an attempt to quantitate it by a membrane-based microwave-mediated electrochemical enzyme-linked immunosorbent assay (MMeELISA) using p-aminophenyl phosphate (pAPP) with an over-all time of 1.5 h. A differential pulse voltammetric signal increased linearly with an increase in the amount of TNF-α with a detection limit of 0.48 pg mL(-1). This bio-sensing platform revealed a significant difference in the TNF-α level between GCF samples from periodontal patients and healthy volunteers.


Subject(s)
Cellulose/analogs & derivatives , Electrochemical Techniques , Enzyme-Linked Immunosorbent Assay , Microwaves , Periodontitis/metabolism , Tumor Necrosis Factor-alpha/analysis , Adult , Cellulose/chemistry , Female , Gingival Crevicular Fluid/chemistry , Humans , Male , Middle Aged
13.
EMBO Rep ; 14(4): 356-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23429341

ABSTRACT

The DNA-binding protein TRF2 is essential for telomere protection and chromosome stability in mammals. We show here that TRF2 expression is activated by the Wnt/ß-catenin signalling pathway in human cancer and normal cells as well as in mouse intestinal tissues. Furthermore, ß-catenin binds to TRF2 gene regulatory regions that are functional in a luciferase transactivating assay. Reduced ß-catenin expression in cancer cells triggers a marked increase in telomere dysfunction, which can be reversed by TRF2 overexpression. We conclude that the Wnt/ß-catenin signalling pathway maintains a level of TRF2 critical for telomere protection. This is expected to have an important role during development, adult stem cell function and oncogenesis.


Subject(s)
Gene Expression Regulation , Telomere Homeostasis , Telomeric Repeat Binding Protein 2/metabolism , Wnt Signaling Pathway , Animals , Binding Sites , Female , Gene Expression , HCT116 Cells , Humans , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telomeric Repeat Binding Protein 2/genetics , Transcriptome , beta Catenin/metabolism
14.
Nucleic Acids Symp Ser (Oxf) ; (52): 499-500, 2008.
Article in English | MEDLINE | ID: mdl-18776472

ABSTRACT

Synthesis of 21 nt siRNAs bearing chemically modified dangling ends with a novel nucleobase was achieved. Evaluation of gene silencing of bcr-abl chimeric gene derived from Philadelphia chromosome by thus obtained chemically modified siRNAs was performed using human leukaemia cell line K-562 and resulted in efficient suppression of the targeted gene. siRNAs whose sense strands were modified with a novel base were found to be more effective than a native siRNA and that siRNAs whose antisense strands were modified with a novel base showed largely decreased silencing effects on the contrary.


Subject(s)
Deoxycytidine/analogs & derivatives , Fusion Proteins, bcr-abl/genetics , RNA Interference , RNA, Small Interfering/chemistry , Deoxycytidine/chemistry , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Small Interfering/chemical synthesis
15.
Nucleic Acids Symp Ser (Oxf) ; (52): 679-80, 2008.
Article in English | MEDLINE | ID: mdl-18776562

ABSTRACT

Oligonucleotides can be covalently linked to peptides composed of any sequence of amino acids by SPFC. The peptides incorporated into the conjugates include nuclear localizing signals (NLS), nuclear export signals (NES), membrane fusion domain of some viral proteins and some designed peptides with amphipathic character. Evaluation of biological properties of DNA-peptide conjugates indicated that (a) the conjugates could bind to target RNA and dsDNA with increased affinity, (b) the conjugates were more resistant to cellular nuclease degradation, (c) the conjugates-RNA hybrids could activate RNase H as effective as native oligonucleotides, (d) the conjugates with fusion peptides showed largely enhanced cellular uptake, (e) the conjugates with NLS could be predominantly delivered into cell nucleus, (f) the conjugates with NES could be localized in cytoplasm. As a result, antisense oligonucleotides conjugated with NLS could inhibit human telomerase in human leukemia cells much more strongly than phosphorothioate oligonucleotides.


Subject(s)
Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Phosphorothioate Oligonucleotides/chemistry , Telomerase/antagonists & inhibitors , Cell Nucleus/metabolism , Humans , Jurkat Cells , Nuclear Export Signals , Nuclear Localization Signals , Oligonucleotides, Antisense/chemical synthesis , Oligonucleotides, Antisense/metabolism , Phosphorothioate Oligonucleotides/chemical synthesis , Phosphorothioate Oligonucleotides/metabolism , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...