Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
J Biol Chem ; 291(11): 5960-5970, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26757822

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is a potential cancer therapy that selectively targets cancer cell death while non-malignant cells remain viable. Using a panel of normal human fibroblasts, we characterized molecular differences in human foreskin fibroblasts and WI-38 TRAIL-resistant cells and marginally sensitive MRC-5 cells compared with TRAIL-sensitive human lung and colon cancer cells. We identified decreased caspase-8 protein expression and protein stability in normal fibroblasts compared with cancer cells. Additionally, normal fibroblasts had incomplete TRAIL-induced caspase-8 activation compared with cancer cells. We found that normal fibroblasts lack the ubiquitin modification of caspase-8 required for complete caspase-8 activation. Treatment with the deubiquitinase inhibitor PR-619 increased caspase-8 ubiquitination and caspase-8 enzymatic activity and sensitized normal fibroblasts to TRAIL-mediated apoptosis. Therefore, posttranslational regulation of caspase-8 confers resistance to TRAIL-induced cell death in normal cells through blockade of initiation of the extrinsic cell death pathway.


Subject(s)
Aminopyridines/pharmacology , Caspase 8/metabolism , Cell Death/drug effects , Fibroblasts/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thiocyanates/pharmacology , Ubiquitination/drug effects , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Humans
2.
Mol Cancer ; 14: 99, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25927855

ABSTRACT

BACKGROUND: We previously reported the identification of ONC201/TIC10, a novel small molecule inducer of the human TRAIL gene that improves efficacy-limiting properties of recombinant TRAIL and is in clinical trials in advanced cancers based on its promising safety and antitumor efficacy in several preclinical models. METHODS: We performed a high throughput luciferase reporter screen using the NCI Diversity Set II to identify TRAIL-inducing compounds. RESULTS: Small molecule-mediated induction of TRAIL reporter activity was relatively modest and the majority of the hit compounds induced low levels of TRAIL upregulation. Among the candidate TRAIL-inducing compounds, TIC9 and ONC201/TIC10 induced sustained TRAIL upregulation and apoptosis in tumor cells in vitro and in vivo. However, ONC201/TIC10 potentiated tumor cell death while sparing normal cells, unlike TIC9, and lacked genotoxicity in normal fibroblasts. Investigating the effects of TRAIL-inducing compounds on cell signaling pathways revealed that TIC9 and ONC201/TIC10, which are the most potent inducers of cell death, exclusively activate Foxo3a through inactivation of Akt/ERK to upregulate TRAIL and its pro-apoptotic death receptor DR5. CONCLUSION: These studies reveal the selective activity of ONC201/TIC10 that led to its selection as a lead compound for this novel class of antitumor agents and suggest that ONC201/TIC10 is a unique inducer of the TRAIL pathway through its concomitant regulation of the TRAIL ligand and its death receptor DR5.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Genes, Reporter , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Imidazoles , Luciferases/metabolism , Mutagens/toxicity , Promoter Regions, Genetic/genetics , Pyridines , Pyrimidines , Small Molecule Libraries/chemistry , Transcription, Genetic/drug effects , Up-Regulation/drug effects
3.
Cancer Cell ; 12(1): 66-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17613437

ABSTRACT

Cells expressing oncogenic c-Myc are sensitized to TNF superfamily proteins. c-Myc also is an important factor in determining whether a cell is sensitive to TRAIL-induced apoptosis, and it is well established that the mitochondrial pathway is essential for apoptosis induced by c-Myc. We investigated whether c-Myc action on the mitochondria is required for TRAIL sensitivity and found that Myc sensitized cells with defective intrinsic signaling to TRAIL. TRAIL induced expression of antiapoptotic Mcl-1 and cIAP2 through activation of NF-kappaB. Both Myc and the multikinase inhibitor sorafenib block NF-kappaB. Combining sorafenib with TRAIL in vivo showed dramatic efficacy in TRAIL-resistant tumor xenografts. We propose the combination of TRAIL with sorafenib holds promise for further development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Benzenesulfonates/pharmacology , Inhibitor of Apoptosis Proteins/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-myc/physiology , Pyridines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Animals , Apoptosis/drug effects , Baculoviral IAP Repeat-Containing 3 Protein , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-bcl-2/genetics , Sorafenib , Transcription, Genetic/physiology , Ubiquitin-Protein Ligases
4.
Clin Chem ; 60(2): 323-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24132944

ABSTRACT

BACKGROUND: The dissemination of circulating tumor cells (CTCs) that cause metastases in distant organs accounts for the majority of cancer-related deaths. CTCs have been established as a cancer biomarker of known prognostic value. The enrichment of viable CTCs for ex vivo analysis could further improve cancer diagnosis and guide treatment selection. We designed a new flexible micro spring array (FMSA) device for the enrichment of viable CTCs independent of antigen expression. METHODS: Unlike previous microfiltration devices, flexible structures at the micro scale minimize cell damage to preserve viability, while maximizing throughput to allow rapid enrichment directly from whole blood with no need for sample preprocessing. Device performance with respect to capture efficiency, enrichment against leukocytes, viability, and proliferability was characterized. CTCs and CTC microclusters were enriched from clinical samples obtained from breast, lung, and colorectal cancer patients. RESULTS: The FMSA device enriched tumor cells with 90% capture efficiency, higher than 10(4) enrichment, and better than 80% viability from 7.5-mL whole blood samples in <10 min on a 0.5-cm(2) device. The FMSA detected at least 1 CTC in 16 out of 21 clinical samples (approximately 76%) compared to 4 out of 18 (approximately 22%) detected with the commercial CellSearch® system. There was no incidence of clogging in over 100 tested fresh whole blood samples. CONCLUSIONS: The FMSA device provides a versatile platform capable of viable enrichment and analysis of CTCs from clinically relevant volumes of whole blood.


Subject(s)
Cell Separation/instrumentation , High-Throughput Screening Assays/instrumentation , Neoplastic Cells, Circulating , Tissue Array Analysis/instrumentation , Cell Count , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Cell Separation/methods , Cell Survival , Equipment Design , High-Throughput Screening Assays/methods , Humans , Leukocytes/cytology , Models, Biological , Neoplastic Cells, Circulating/pathology , Tissue Array Analysis/methods
5.
Am J Pathol ; 179(5): 2169-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21975022

ABSTRACT

Bone marrow-derived cells (BMDCs) participate in the growth and spread of tumors of the breast, brain, lung, and stomach. To date, there are limited reports of bone marrow involvement in colon cancer pathogenesis, but such findings would have the potential to generate novel treatments for colon cancer patients. We have established a mouse model for imaging BMDCs from whole tumor to single-cell resolution, whereby the bone marrow of lethally irradiated host animals is reconstituted with EGFP-expressing bone marrow cells from matched TgActb(EGFP) donors. The BM transplants yield mice with fluorescently labeled bone marrow, and so BMDCs can subsequently be monitored within a tumor through optical imaging. Successful BM reconstitution was confirmed at 8 weeks after transplantation, when surviving BALB/c mice were injected with CT26 mouse colon cancer cells. We find that up to 45% of cells dissociated from the tumors are GFP(+) and approximately 50% of Lin(+), CD11b(+), and CD3(+) cells express high levels of GFP. Notably, tumor growth is reduced in BM transplanted animals, compared with untransplanted host mice or EGFP-expressing BM donor mice. A needed next step is to separate the molecular and cellular (eg, T cells, NK cells, macrophages) bases of the antitumor effect of the BMDCs from any protumorigenic effect that could be subverted for therapeutic gain.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Colonic Neoplasms/therapy , Green Fluorescent Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Colonic Neoplasms/pathology , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Transplantation/methods , Transplantation, Isogeneic/methods , Tumor Microenvironment , Whole-Body Irradiation
6.
Cancer Cell ; 6(6): 597-609, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15607964

ABSTRACT

p53-dependent apoptosis is a major determinant of its tumor suppressor activity and can be triggered by hypoxia. No p53 target is known to be induced by p53 or to mediate p53-dependent apoptosis during hypoxia. We report that p53 can directly upregulate expression of Bnip3L, a cell death inducer. During hypoxia, Bnip3L is highly induced in wild-type p53-expressing cells, in part due to increased recruitment of p53 and CBP to Bnip3L. Apoptosis is reduced in hypoxia-exposed cells with functional p53 following Bnip3L knockdown. In vivo, Bnip3L knockdown promotes tumorigenicity of wild-type versus mutant p53-expressing tumors. Thus, Bnip3L, capable of attenuating tumorigenicity, mediates p53-dependent apoptosis under hypoxia, which provides a novel understanding of p53 in tumor suppression.


Subject(s)
Membrane Proteins/physiology , Neoplasms/pathology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , CREB-Binding Protein , Caspase 3 , Caspases/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Cisplatin/pharmacology , Doxycycline/pharmacology , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/genetics , Glucose Transporter Type 1 , Humans , Immunohistochemistry , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Monosaccharide Transport Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Trans-Activators/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
7.
Am J Cancer Res ; 12(1): 315-326, 2022.
Article in English | MEDLINE | ID: mdl-35141020

ABSTRACT

TRAIL-based therapies are of significant clinical interest because of its unique ability to induce apoptosis in cancer cells while sparing normal and untransformed cells. This selective antitumor potential of the TRAIL pathway has been harnessed by development of therapeutics including recombinant (rh)TRAIL and TRAIL-receptor agonist antibodies such as mapatumumab and lexatumumab. While these TRAIL-based therapies have proven successful in preclinical studies and safe in early phase clinical trials, the limited serum half-life has been a hurdle for further clinical development. Here we characterize miR-3132, a novel and first-in class TRAIL-inducing miRNA with potent anti-proliferative and pro-apoptotic effects in cancer cell lines. Initial mechanistic studies indicate that miR-3132 engages the interferon signaling pathway to induce TRAIL and subsequent TRAIL-dependent apoptosis in cancer cell lines. Our data further suggests that the binding of miR-3132 to toll-like receptors could be the upstream pathway for the interferon response. The current study the first report to demonstrate miR-3132's in vitro efficacy and preliminary mechanism of action in cancer cell lines.

8.
Sci Rep ; 11(1): 20871, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34686682

ABSTRACT

A prevalent characteristic of solid tumors is intra-tumoral hypoxia. Hypoxia-inducible factor 1α (HIF1α) predominantly mediates the adaptive response to O2 oscillation and is linked to multiple malignant hallmarks. Here we describe a strategy to robustly target HIF1α by dual inhibition of CDK(s) and heat shock protein 90 (HSP90). We show that CDK1 may contribute to HSP90-mediated HIF1α stabilization. CDK1 knockdown enhances the decrease of HIF1α by HSP90 inhibition. Dual inhibition of CDK1 and HSP90 significantly increases apoptosis and synergistically inhibits cancer cell viability. Similarly, targeting CDK4/6 using FDA-approved inhibitors in combination with HSP90 inhibition shows a class effect on HIF1α inhibition and cancer cell viability suppression not only in colorectal but also in various other cancer types, including Rb-deficient cancer cells. Dual inhibition of CDK4/6 and HSP90 suppresses tumor growth in vivo. In summary, combined targeting of CDK(s) (CDK1 or CDK4/6) and HSP90 remarkably inhibits the expression level of HIF1α and shows promising anti-cancer efficacy with therapeutic potential.


Subject(s)
Cyclin-Dependent Kinases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proteasome Endopeptidase Complex/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , HCT116 Cells , HT29 Cells , Humans , Hypoxia/metabolism
9.
Cancer Biol Ther ; 22(10-12): 554-563, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34696710

ABSTRACT

ONC201 demonstrated promising activity in patients with advanced endometrial cancer in a Phase I clinical trial. ONC201 activates the integrated stress response (ISR) and upregulates TRAIL and its receptor DR5. We hypothesized ONC201 upregulation of DR5 could sensitize tumors to TRAIL and combination of ONC201 and TRAIL would lead to enhanced cell death in endometrial cancer models. Five endometrial cancer cell lines AN3CA, HEC1A, Ishikawa, RL952, and KLE as well as a murine xenograft model were treated with ONC201 alone or in combination with TRAIL. ONC201 decreased the cell viability of all five endometrial cancer cell lines at clinically achievable low micro-molar concentrations (2-4 µM). ONC201 activated the ISR and induced protein expression of TRAIL and DR5 at the cell surface. Pretreatment with ONC201 sensitized endometrial cancer cell lines to TRAIL, leading to increased cell death induction compared to either agent alone. Tumor growth was reduced in vivo by the ONC201/TRAIL combination treatment in the xenograft model of endometrial cancer (p = .014). Mice treated with combination treatment survived significantly longer than mice from the three control groups (p = .018). ONC201 decreased cell viability in endometrial cancer cells lines primarily through growth arrest while the combination of ONC201 and TRAIL promoted cell death in vitro and in vivo. Our results suggest a novel cancer therapeutic strategy that can be further investigated in the clinic.


Subject(s)
Antineoplastic Agents , Endometrial Neoplasms , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Female , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Imidazoles , Mice , Pyridines/pharmacology , Pyrimidines , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand
10.
Elife ; 102021 07 29.
Article in English | MEDLINE | ID: mdl-34324416

ABSTRACT

Mutations in TP53 occur commonly in the majority of human tumors and confer aggressive tumor phenotypes, including metastasis and therapy resistance. CB002 and structural-analogs restore p53 signaling in tumors with mutant-p53 but we find that unlike other xanthines such as caffeine, pentoxifylline, and theophylline, they do not deregulate the G2 checkpoint. Novel CB002-analogs induce pro-apoptotic Noxa protein in an ATF3/4-dependent manner, whereas caffeine, pentoxifylline, and theophylline do not. By contrast to caffeine, CB002-analogs target an S-phase checkpoint associated with increased p-RPA/RPA2, p-ATR, decreased Cyclin A, p-histone H3 expression, and downregulation of essential proteins in DNA-synthesis and DNA-repair. CB002-analog #4 enhances cell death, and decreases Ki-67 in patient-derived tumor-organoids without toxicity to normal human cells. Preliminary in vivo studies demonstrate anti-tumor efficacy in mice. Thus, a novel class of anti-cancer drugs shows the activation of p53 pathway signaling in tumors with mutated p53, and targets an S-phase checkpoint.


Subject(s)
Aniline Compounds/pharmacology , Mutation , Purines/pharmacology , S Phase Cell Cycle Checkpoints/genetics , Signal Transduction/drug effects , Transcriptome , Tumor Suppressor Protein p53/genetics , Aniline Compounds/chemistry , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage , Female , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Purines/chemistry , Purines/therapeutic use , Random Allocation , Xenograft Model Antitumor Assays
11.
Neoplasia ; 23(3): 304-325, 2021 03.
Article in English | MEDLINE | ID: mdl-33582407

ABSTRACT

A long-term goal in the cancer-field has been to develop strategies for treating p53-mutated tumors. A novel small-molecule, PG3-Oc, restores p53 pathway-signaling in tumor cells with mutant-p53, independently of p53/p73. PG3-Oc partially upregulates the p53-transcriptome (13.7% of public p53 target-gene dataset; 15.2% of in-house dataset) and p53-proteome (18%, HT29; 16%, HCT116-p53-/-). Bioinformatic analysis indicates critical p53-effectors of growth-arrest (p21), apoptosis (PUMA, DR5, Noxa), autophagy (DRAM1), and metastasis-suppression (NDRG1) are induced by PG3-Oc. ERK1/2- and CDK9-kinases are required to upregulate ATF4 by PG3-Oc which restores p53 transcriptomic-targets in cells without functional-p53. PG3-Oc represses MYC (ATF4-independent), and upregulates PUMA (ATF4-dependent) in mediating cell death. With largely nonoverlapping transcriptomes, induced-ATF4 restores p53 transcriptomic targets in drug-treated cells including functionally important mediators such as PUMA and DR5. Our results demonstrate novel p53-independent drug-induced molecular reprogramming involving ERK1/2, CDK9, and ATF4 to restore upregulation of p53 effector genes required for cell death and tumor suppression.


Subject(s)
Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cyclin-Dependent Kinase 9/metabolism , Mutation , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , CRISPR-Cas Systems , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum Stress , Gene Editing , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System , Models, Biological
13.
Oncotarget ; 11(42): 3753-3769, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33144917

ABSTRACT

ONC201 was initially identified as an inducer of cell death through the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway. The compound is currently being tested in patients with hematological malignancies and solid tumors, including those of the breast. We investigated strategies to convert the response of breast cancers to ONC201 from anti-proliferative to apoptotic. ONC201 treatment upregulates TRAIL and primes TRAIL-resistant non-triple negative breast cancer (TNBC) cells to undergo cell death through the extrinsic pathway. Remarkably, the addition of exogenous recombinant human TRAIL (rhTRAIL) converts the response of TRAIL-resistant non-TNBC cells to ONC201 from anti-proliferative to apoptotic in a death receptor 5 (DR5)-dependent manner in vitro. Importantly, normal fibroblasts do not undergo apoptosis following rhTRAIL plus ONC201. In vivo, MDA-MB-361 tumor growth rate is significantly reduced following treatment with a combination of ONC201 and rhTRAIL as compared to control tumors. Natural killer (NK) cells which use TRAIL to kill DR5-expressing cancer cells, exhibit greater cytotoxicity against ONC201-treated breast cancer cells compared to controls. rhTRAIL also converts the response of cells from other tumor types to ONC201 from anti-proliferative to apoptotic. A monoclonal DR5-agonistic antibody converts the response of non-TNBC cells to ONC201 from anti-proliferative to apoptotic. Our findings describe a novel therapeutic strategy that potently converts the response of a cancer cell to ONC201 from anti-proliferative to apoptotic. This approach may be clinically relevant and has potential to induce tumor regression of patient tumors with relative resistance to ONC201 monotherapy.

14.
Cancer Res ; 67(19): 9443-54, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909054

ABSTRACT

The antineoplastic drug sorafenib (BAY 43-9006) is a multikinase inhibitor that targets the serine-threonine kinase B-Raf as well as several tyrosine kinases. Given the numerous molecular targets of sorafenib, there are several potential anticancer mechanisms of action, including induction of apoptosis, cytostasis, and antiangiogenesis. We observed that sorafenib has broad activity in viability assays in several human tumor cell lines but selectively induces apoptosis in only some lines. Sorafenib was found to decrease Mcl-1 levels in most cell lines tested, but this decrease did not correlate with apoptotic sensitivity. Sorafenib slows cell cycle progression and prevents irradiated cells from reaching and accumulating at G2-M. In synchronized cells, sorafenib causes a reversible G1 delay, which is associated with decreased levels of cyclin D1, Rb, and phosphorylation of Rb. Although sorafenib does not affect intrinsic radiosensitivity using in vitro colony formation assays, it significantly reduces colony size. In HCT116 xenograft tumor growth delay experiments in mice, sorafenib alters radiation response in a schedule-dependent manner. Radiation treatment followed sequentially by sorafenib was found to be associated with the greatest tumor growth delay. This study establishes a foundation for clinical testing of sequential fractionated radiation followed by sorafenib in gastrointestinal and other malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Pyridines/pharmacology , Animals , Cell Division/drug effects , Cell Division/radiation effects , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Combined Modality Therapy , Cyclin D1/blood , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Dose-Response Relationship, Radiation , Drug Administration Schedule , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Female , G2 Phase/drug effects , G2 Phase/radiation effects , HCT116 Cells , Humans , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/metabolism , Sorafenib , Xenograft Model Antitumor Assays
15.
Cell Cycle ; 17(5): 557-567, 2018.
Article in English | MEDLINE | ID: mdl-28749203

ABSTRACT

P53 tumor suppressor gene mutations occur in the majority of human cancers and contribute to tumor development, progression and therapy resistance. Direct functional restoration of p53 as a transcription factor has been difficult to achieve in the clinic. We performed a functional screen using a bioluminescence-based transcriptional read-out to identify small molecules that restore the p53 pathway in mutant p53-bearing cancer cells. We identified CB002, as a candidate that restores p53 function in mutant p53-expressing colorectal cancer cells and without toxicity to normal human fibroblasts. Cells exposed to CB002 show increased expression of endogenous p53 target genes NOXA, DR5, and p21 and cell death which occurs by 16 hours, as measured by cleaved caspases or PARP. Stable knockdown of NOXA completely abrogates PARP cleavage and reduces sub-G1 content, implicating NOXA as the key mediator of cell death induction by CB002. Moreover, CB002 decreases the stability of mutant p53 in RXF393 cancer cells and an exogenously expressed R175H p53 mutant in HCT116 p53-null cells. R175H p53 expression was rescued by addition of proteasome inhibitor MG132 to CB002, suggesting a role for ubiquitin-mediated degradation of the mutant protein. In summary, CB002, a p53 pathway-restoring compound that targets mutant p53 for degradation and induces tumor cell death through NOXA, may be further developed as a cancer therapeutic.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Purines/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Leupeptins/pharmacology , Mutagenesis, Site-Directed , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics
16.
Mol Cancer Res ; 16(5): 754-766, 2018 05.
Article in English | MEDLINE | ID: mdl-29588330

ABSTRACT

Androgen receptor (AR) signaling plays a key role in prostate cancer progression, and androgen deprivation therapy (ADT) is a mainstay clinical treatment regimen for patients with advanced disease. Unfortunately, most prostate cancers eventually become androgen-independent and resistant to ADT with patients progressing to metastatic castration-resistant prostate cancer (mCRPC). Constitutively activated AR variants (AR-V) have emerged as mediators of resistance to AR-targeted therapy and the progression of mCRPC, and they represent an important therapeutic target. Out of at least 15 AR-Vs described thus far, AR-V7 is the most abundant, and its expression correlates with ADT resistance. ONC201/TIC10 is the founding member of the imipridone class of small molecules and has shown anticancer activity in a broad range of tumor types. ONC201 is currently being tested in phase I/II clinical trials for advanced solid tumors, including mCRPC, and hematologic malignancies. There has been promising activity observed in patients in early clinical testing. This study demonstrates preclinical single-agent efficacy of ONC201 using in vitro and in vivo models of prostate cancer. ONC201 has potent antiproliferative and proapoptotic effects in both castration-resistant and -sensitive prostate cancer cells. Furthermore, the data demonstrate that ONC201 downregulates the expression of key drivers of prostate cancer such as AR-V7 and downstream target genes including the clinically used biomarker PSA (KLK3). Finally, the data also provide a preclinical rationale for combination of ONC201 with approved therapeutics for prostate cancer such as enzalutamide, everolimus (mTOR inhibitor), or docetaxel.Implications: The preclinical efficacy of ONC201 as a single agent or in combination, in hormone-sensitive or castration-resistant prostate cancer, suggests the potential for immediate clinical translation. Mol Cancer Res; 16(5); 754-66. ©2018 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Everolimus/therapeutic use , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Everolimus/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Imidazoles , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Pyridines , Pyrimidines , Signal Transduction , Transfection
17.
Neoplasia ; 20(1): 80-91, 2018 01.
Article in English | MEDLINE | ID: mdl-29216597

ABSTRACT

ONC201/TIC10 is a first-in-class small molecule inducer of TRAIL that causes early activation of the integrated stress response. Its promising safety profile and broad-spectrum efficacy in vitro have been confirmed in Phase I/II trials in several advanced malignancies. Binding and reporter assays have shown that ONC201 is a selective antagonist of the dopamine D2-like receptors, specifically, DRD2 and DRD3. We hypothesized that ONC201's interaction with DRD2 plays a role in ONC201's anticancer effects. Using cBioportal and quantitative reverse-transcription polymerase chain reaction analyses, we confirmed that DRD2 is expressed in different cancer cell types in a cell type-specific manner. On the other hand, DRD3 was generally not detectable. Overexpressing DRD2 in cells with low DRD2 levels increased ONC201-induced PARP cleavage, which was preceded and correlated with an increase in ONC201-induced CHOP mRNA expression. On the other hand, knocking out DRD2 using CRISPR/Cas9 in three cancer cell lines was not sufficient to abrogate ONC201's anticancer effects. Although ONC201's anticancer activity was not dependent on DRD2 expression in the cancer cell types tested, we assessed the cytotoxic potential of DRD2 blockade. Transient DRD2 knockdown in HCT116 cells activated the integrated stress response and reduced cell number. Pharmacological antagonism of DRD2 significantly reduced cell viability. Thus, we demonstrate in this study that disrupting dopamine receptor expression and activity can have cytotoxic effects that may at least be in part due to the activation of the integrated stress response. On the other hand, ONC201's anticancer activity goes beyond its ability to antagonize DRD2, potentially due to ONC201's ability to activate other pathways that are independent of DRD2. Nevertheless, blocking the dopamine D1-like receptor DRD5 via siRNA or the use of a pharmacological antagonist promoted ONC201-induced anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplasms/metabolism , Receptors, Dopamine/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Imidazoles , Neoplasms/genetics , Pyridines , Pyrimidines , RNA, Small Interfering/genetics , Receptors, Dopamine/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism
18.
Cell Cycle ; 17(4): 468-478, 2018.
Article in English | MEDLINE | ID: mdl-29157092

ABSTRACT

ONC201, founding member of the imipridone class of small molecules, is currently being evaluated in advancer cancer clinical trials. We explored single agent and combinatorial efficacy of ONC201 in preclinical models of hematological malignancies. ONC201 demonstrated (GI50 1-8 µM) dose- and time-dependent efficacy in acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt's lymphoma, anaplastic large cell lymphoma (ALCL), cutaneous T-cell lymphoma (CTCL), Hodgkin's lymphoma (nodular sclerosis) and multiple myeloma (MM) cell lines including cells resistant to standard of care (dexamethasone in MM) and primary samples. ONC201 induced caspase-dependent apoptosis that involved activation of the integrated stress response (ATF4/CHOP) pathway, inhibition of Akt phosphorylation, Foxo3a activation, downregulation of cyclin D1, IAP and Bcl-2 family members. ONC201 synergistically reduced cell viability in combination with cytarabine and 5-azacytidine in AML cells. ONC201 combined with cytarabine in a Burkitt's lymphoma xenograft model induced tumor growth inhibition that was superior to either agent alone. ONC201 synergistically combined with bortezomib in MM, MCL and ALCL cells and with ixazomib or dexamethasone in MM cells. ONC201 combined with bortezomib in a Burkitt's lymphoma xenograft model reduced tumor cell density and improved CHOP induction compared to either agent alone. These results serve as a rationale for ONC201 single-agent trials in relapsed/refractory acute leukemia, non-Hodgkin's lymphoma, MM and combination trial with dexamethasone in MM, provide pharmacodynamic biomarkers and identify further synergistic combinatorial regimens that can be explored in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Activating Transcription Factor 4/metabolism , Animals , Antineoplastic Agents/therapeutic use , Azacitidine/pharmacology , Boron Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , G1 Phase Cell Cycle Checkpoints/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Imidazoles , Mice , Mice, SCID , Pyridines , Pyrimidines , Transcription Factor CHOP/metabolism , Transplantation, Heterologous
19.
Cancer Biol Ther ; 6(3): 447-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17387267

ABSTRACT

Molecular imaging techniques can detect and monitor characteristics of the tumor microenvironment, such as angiogenesis, hypoxia, metabolism, and apoptosis that may better correlate with response to cancer therapy and may provide information in real-time. We investigated the use of a novel, spatially discrete, hyperspectral, multi-fiber optical system to characterize selected regions of skin in living mice. We determined the reproducibility and robustness of the spectral signatures derived from comparable regions of interest. Additionally, we characterized spectral differences in vascular and non-vascular fields to determine their potential use in monitoring angiogenesis. The macroscopic Prism and Reflectance Imaging Spectroscopy System (MACRO-PARISS) was calibrated against a National Institute for Standards and Technology (NIST)-certified lamp, allowing for reproducible spectra with any instrument similarly calibrated. Spectra were classified using a linearity-independent algorithm over a wavelength range of 450-920 nm. Classified spectra were integrated into a spectral library and subsequent acquisitions were correlated with the library set to a minimum correlation coefficient (MCC) of 99%. The results indicated that similar regions of interest with respect to vascularity consistently generated a unique spectral signature. As the field of view (FOV) moved from vascular to non-vascular areas, the acquired spectra changed in a step-wise and predictable fashion. Additionally, vascular fields that were deprived of their blood supply subsequently generated a non-vascular spectral signature. This work has implications for the monitoring of various physiologic or pathological processes including tumor angiogenesis and the therapeutic effects of anti-vascular agents.


Subject(s)
Fiber Optic Technology/instrumentation , Monitoring, Physiologic/instrumentation , Neovascularization, Physiologic , Skin/blood supply , Spectrum Analysis/methods , Animals , Mice , Mice, SCID , Reproducibility of Results
20.
Mol Cell Biol ; 23(16): 5556-71, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12897130

ABSTRACT

Loss of p53 sensitizes to antimicrotubule agents in human tumor cells, but little is known about its role during mitosis. We have identified the Polo-like kinase family member serum inducible kinase (Snk/Plk2) as a novel p53 target gene. Snk/Plk2 mutagenesis demonstrated that its kinase activity is negatively regulated by its C terminus. Small interfering RNA (siRNA)-mediated Snk/Plk2 silencing in the presence of the mitotic poisons paclitaxel (Taxol) or nocodazole significantly increased apoptosis, similar to p53 mutations, which confer paclitaxel sensitivity. Furthermore, we have demonstrated that the apoptosis due to silencing of Snk/Plk2 in the face of spindle damage occurs in mitotic cells and not in cells that have progressed to a G(1)-like state without dividing. Since siRNA directed against Snk/Plk2 promoted death of paclitaxel-treated cells in mitosis, we envision a mitotic checkpoint wherein p53-dependent activation of Snk/Plk2 prevents mitotic catastrophe following spindle damage. Finally, these studies suggest that disruption of Snk/Plk2 may be of therapeutic value in sensitizing paclitaxel-resistant tumors.


Subject(s)
Gene Silencing , Genes, p53 , Mitosis , Paclitaxel/pharmacology , Serine Endopeptidases/genetics , Transcription Factors/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Base Sequence , Blotting, Northern , Blotting, Western , Cell Death , Cell Line , Chromatin/metabolism , DNA Damage , Dose-Response Relationship, Radiation , Female , Flow Cytometry , G1 Phase , Green Fluorescent Proteins , HeLa Cells , Humans , In Situ Hybridization , Luciferases/metabolism , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microtubules/drug effects , Models, Biological , Plasmids/metabolism , Precipitin Tests , Protein Structure, Tertiary , RNA/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Temperature , Time Factors , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL