Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Methods Mol Biol ; 2318: 337-346, 2021.
Article in English | MEDLINE | ID: mdl-34019301

ABSTRACT

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of the Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Subject(s)
Drug Screening Assays, Antitumor/methods , Genes, myc/drug effects , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Cell Line, Tumor , Genes, myc/genetics , Genes, myc/physiology , Humans , Oncogene Proteins/drug effects , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Small Molecule Libraries/pharmacology , Transcription Factors/drug effects , Transcription Factors/metabolism
2.
Commun Biol ; 4(1): 466, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846531

ABSTRACT

The Toll-like receptor 5 (TLR5) agonist entolimod, a derivative of Salmonella flagellin, has therapeutic potential for several indications including radioprotection and cancer immunotherapy. However, in Phase 1 human studies, entolimod induced a rapid neutralizing immune response, presumably due to immune memory from prior exposure to flagellated enterobacteria. To enable multi-dose applications, we used structure-guided reengineering to develop a next-generation, substantially deimmunized entolimod variant, GP532. GP532 induces TLR5-dependent NF-κB activation like entolimod but is smaller and has mutations eliminating an inflammasome-activating domain and key B- and T-cell epitopes. GP532 is resistant to human entolimod-neutralizing antibodies and shows reduced de novo immunogenicity. GP532 also has improved bioavailability, a stronger effect on key cytokine biomarkers, and a longer-lasting effect on NF-κB. Like entolimod, GP532 demonstrated potent prophylactic and therapeutic efficacy in mouse models of radiation-induced death and tissue damage. These results establish GP532 as an optimized TLR5 agonist suitable for multi-dose therapies and for patients with high titers of preexisting flagellin-neutralizing antibodies.


Subject(s)
Peptides/pharmacology , Signal Transduction , Toll-Like Receptor 5/agonists , Cell Line, Tumor , HEK293 Cells , Humans
3.
Neuro Oncol ; 19(2): 186-196, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27370399

ABSTRACT

Background: The survival rate for patients with glioblastoma (GBM) remains dismal. New therapies targeting molecular pathways dysregulated in GBM are needed. One such clinical-stage drug candidate, CBL0137, is a curaxin, small molecules which simultaneously downregulate nuclear factor-kappaB (NF-ĸB) and activate p53 by inactivating the chromatin remodeling complex, Facilitates Chromatin Transcription (FACT). Methods: We used publicly available databases to establish levels of FACT subunit expression in GBM. In vitro, we evaluated the toxicity and effect of CBL0137 on FACT, p53, and NF-ĸB on U87MG and A1207 human GBM cells. In vivo, we implanted the cells orthotopically in nude mice and administered CBL0137 in various dosing regimens to assess brain and tumor accumulation of CBL0137, its effect on tumor cell proliferation and apoptosis, and on survival of mice with and without temozolomide (TMZ). Results: FACT subunit expression was elevated in GBM compared with normal brain. CBL0137 induced loss of chromatin-unbound FACT, activated p53, inhibited NF-ĸB-dependent transcription, and was toxic to GBM cells. The drug penetrated the blood-brain barrier and accumulated in orthotopic tumors significantly more than normal brain tissue. It increased apoptosis and suppressed proliferation in both U87MG and A1207 tumors. Intravenous administration of CBL0137 significantly increased survival in models of early- through late-stage TMZ-responsive and -resistant GBM, with a trend toward significantly increasing the effect of TMZ in TMZ-responsive U87MG tumors. Conclusion: CBL0137 targets GBM according to its proposed mechanism of action, crosses the blood-brain barrier, and is efficacious in both TMZ-responsive and -resistant orthotopic models, making it an attractive new therapy for GBM.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Carbazoles/therapeutic use , DNA-Binding Proteins/antagonists & inhibitors , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , High Mobility Group Proteins/antagonists & inhibitors , Transcriptional Elongation Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blood-Brain Barrier , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Res ; 76(22): 6620-6630, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27680682

ABSTRACT

Isolated limb perfusion (ILP) with the chemotherapeutic agent melphalan is an effective treatment option for extremity in-transit melanoma but is toxic and technically challenging to deliver locoregionally. CBL0137 is an experimental clinical drug with broad anticancer activity in animal models, owing to its ability to bind DNA in a nongenotoxic manner and inactivate the FACT chromatin modulator essential for tumor cell viability. Here, we report that CBL0137 delivered by ILP in a murine melanoma model is as efficacious as melphalan, displaying antitumor activity at doses corresponding to only a fraction of the systemic MTD of CBL0137. The ability to bind DNA quickly combined with a favorable safety profile made it possible to substitute CBL0137 in the ILP protocol, using an intra-arterial infusion method, to safely achieve effective tumor suppression. Our findings of a preclinical proof of concept for CBL0137 and its administration via intra-arterial infusion as a superior treatment compared with melphalan ILP allows for locoregional treatment anywhere a catheter can be placed. Cancer Res; 76(22); 6620-30. ©2016 AACR.


Subject(s)
Extremities/pathology , Infusion Pumps , Melanoma/drug therapy , Animals , Female , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Treatment Outcome , Validation Studies as Topic
5.
Methods Mol Biol ; 1012: 255-64, 2013.
Article in English | MEDLINE | ID: mdl-24006071

ABSTRACT

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Cell Line, Tumor , Gene Expression , Genes, Reporter , Humans , Phenotype , Small Molecule Libraries
6.
Cancer Res ; 72(4): 845-53, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22202125

ABSTRACT

Amplification of the transcription factor MYCN is associated with poor outcome and a multidrug-resistant phenotype in neuroblastoma. N-Myc regulates the expression of several ATP-binding cassette (ABC) transporter genes, thus affecting global drug efflux. Because these transporters do not confer resistance to several important cytotoxic agents used to treat neuroblastoma, we explored the prognostic significance and transcriptional regulation of the phase II detoxifying enzyme, glutathione S-transferase P1 (GSTP1). Using quantitative real-time PCR, GSTP1 gene expression was assessed in a retrospective cohort of 51 patients and subsequently in a cohort of 207 prospectively accrued primary neuroblastomas. These data along with GSTP1 expression data from an independent microarray study of 251 neuroblastoma samples were correlated with established prognostic indicators and disease outcome. High levels of GSTP1 were associated with decreased event-free and overall survival in all three cohorts. Multivariable analyses, including age at diagnosis, tumor stage, and MYCN amplification status, were conducted on the two larger cohorts, independently showing the prognostic significance of GSTP1 expression levels in this setting. Mechanistic investigations revealed that GSTP1 is a direct transcriptional target of N-Myc in neuroblastoma cells. Together, our findings reveal that N-Myc regulates GSTP1 along with ABC transporters that act to control drug metabolism and efflux. Furthermore, they imply that strategies to jointly alter these key multidrug resistance mechanisms may have therapeutic implications to manage neuroblastomas and other malignancies driven by amplified Myc family genes.


Subject(s)
Gene Expression Regulation, Neoplastic , Glutathione S-Transferase pi/metabolism , Neuroblastoma/enzymology , Nuclear Proteins/physiology , Oncogene Proteins/physiology , Animals , Cohort Studies , Female , Humans , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/mortality , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prognosis
7.
Sci Transl Med ; 3(95): 95ra74, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832239

ABSTRACT

Effective eradication of cancer requires treatment directed against multiple targets. The p53 and nuclear factor κB (NF-κB) pathways are dysregulated in nearly all tumors, making them attractive targets for therapeutic activation and inhibition, respectively. We have isolated and structurally optimized small molecules, curaxins, that simultaneously activate p53 and inhibit NF-κB without causing detectable genotoxicity. Curaxins demonstrated anticancer activity against all tested human tumor xenografts grown in mice. We report here that the effects of curaxins on p53 and NF-κB, as well as their toxicity to cancer cells, result from "chromatin trapping" of the FACT (facilitates chromatin transcription) complex. This FACT inaccessibility leads to phosphorylation of the p53 Ser(392) by casein kinase 2 and inhibition of NF-κB-dependent transcription, which requires FACT activity at the elongation stage. These results identify FACT as a prospective anticancer target enabling simultaneous modulation of several pathways frequently dysregulated in cancer without induction of DNA damage. Curaxins have the potential to be developed into effective and safe anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Transcriptional Elongation Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Carbazoles/chemistry , Casein Kinase II/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/metabolism , Cisplatin/pharmacology , DNA Damage , Humans , Mice , Models, Biological , NF-kappa B/metabolism , Protein Binding/drug effects , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
8.
Cancer Res ; 69(16): 6573-80, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19654298

ABSTRACT

The multidrug resistance-associated protein 1 (MRP1) has been closely linked to poor treatment response in several cancers, most notably neuroblastoma. Homozygous deletion of the MRP1 gene in primary murine neuroblastoma tumors resulted in increased sensitivity to MRP1 substrate drugs (vincristine, etoposide, and doxorubicin) compared with tumors containing both copies of wild-type MRP1, indicating that MRP1 plays a significant role in the drug resistance in this tumor type and defining this multidrug transporter as a target for pharmacologic suppression. A cell-based readout system was created to functionally determine intracellular accumulation of MRP1 substrates using a p53-responsive reporter as an indicator of drug-induced DNA damage. Screening of small-molecule libraries in this readout system revealed pyrazolopyrimidines as a prominent structural class of potent MRP1 inhibitors. Reversan, the lead compound of this class, increased the efficacy of both vincristine and etoposide in murine models of neuroblastoma (syngeneic and human xenografts). As opposed to the majority of inhibitors of multidrug transporters, Reversan was not toxic by itself nor did it increase the toxicity of chemotherapeutic drug exposure in mice. Therefore, Reversan represents a new class of nontoxic MRP1 inhibitor, which may be clinically useful for the treatment of neuroblastoma and other MRP1-overexpressing drug-refractory tumors by increasing their sensitivity to conventional chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neuroblastoma/drug therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Animals , Dogs , Drug Synergism , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Multidrug Resistance-Associated Proteins/genetics , Small Molecule Libraries/analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 101(34): 12664-9, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15314226

ABSTRACT

The mechanisms causing persistence of embryonal cells that later give rise to tumors is unknown. One tumorigenic factor in the embryonal childhood tumor neuroblastoma is the MYCN protooncogene. Here we show that normal mice developed neuroblast hyperplasia in paravertebral ganglia at birth that completely regressed by 2 weeks of age. In contrast, ganglia from MYCN transgenic (TH-MYCN) mice demonstrated a marked increase in neuroblast hyperplasia and MycN expression during week 1. Regression of neuroblast hyperplasia was then delayed and incomplete before neuroblastoma tumor formation at 6 and 13 weeks in homo- and hemizygote mice, respectively. Paravertebral neuronal cells cultured from perinatal TH-MYCN mice exhibited 3- to 10-fold resistance to nerve growth factor (NGF) withdrawal, compared with normal mice. Both low- and high-affinity NGF receptors were expressed in perinatal neuroblast hyperplasia but not in neuroblastoma tumor tissue. MYCN transgene amplification was present at low levels in perinatal neuroblast hyperplasia from both homo- and hemizygote TH-MYCN mice. However, only in hemizygous mice did tumor formation correlate with a stepwise increase in the frequency of MYCN amplification. These data suggest that inappropriate perinatal MycN expression in paravertebral ganglia cells from TH-MYCN mice initiated tumorigenesis by altering the physiologic process of neural crest cell deletion. Persisting embryonal neural crest cells underwent further changes, such as MYCN amplification and repression of NGF receptor expression, during tumor progression. Our studies provide a model for studying perinatal factors influencing embryonal tumor initiation.


Subject(s)
Carcinoma, Embryonal/metabolism , Gene Amplification , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Animals , Apoptosis/physiology , Carcinoma, Embryonal/pathology , Culture Techniques , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/metabolism , Ganglia, Sympathetic/pathology , Humans , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Nerve Growth Factor/metabolism , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Receptors, Nerve Growth Factor/metabolism , Transgenes
10.
J Natl Cancer Inst ; 95(18): 1394-403, 2003 Sep 17.
Article in English | MEDLINE | ID: mdl-13130115

ABSTRACT

BACKGROUND: Human MYCN (hMYCN) oncogene amplification is a powerful predictor of treatment failure in childhood neuroblastoma, and dysregulation of hMYCN protein expression appears to be critically involved in the pathogenesis of this disease. We used hMYCN antisense (AS) oligonucleotides to investigate, both in vitro and in vivo, the therapeutic potential of inhibiting hMYCN expression. METHODS: We transiently transfected human neuroblastoma IMR-32 cells, which have an amplified hMYCN gene, with fluorescently labeled hMYCN AS or scrambled (SCR) control oligonucleotides and used fluorescence-activated cell sorting to enrich for cell populations containing different levels of the oligonucleotides. We used fluorescence immunocytochemistry or reverse transcription polymerase chain reaction to assay gene expression levels and trypan blue exclusion to assay growth inhibition in the cell populations. We examined the effects of continuous treatment for 6 weeks with AS or SCR oligonucleotides via subcutaneously implanted microosmotic pumps on tumor growth in a transgenic mouse model of hMYCN-induced neuroblastoma (n = 20 mice per group). All statistical tests were two-sided. RESULTS: IMR-32 cells treated with AS oligonucleotides had approximately half as much hMYCN protein and cell proliferation as either SCR oligonucleotide-transfected or mock-transfected controls; the differences were statistically significant. Transgenic mice treated with AS oligonucleotides had lower tumor incidence and statistically significantly lower tumor mass than SCR-treated or untreated control mice. Compared with control treatments, AS oligonucleotide treatment in vitro and in vivo was associated with decreased expression of hMYCN and putative hMYCN target genes but not with that of closely related genes. Several AS oligonucleotide-treated mice developed tumors contralateral to the site of oligonucleotide administration, whereas SCR oligonucleotide-treated or untreated mice displayed bilateral tumor growth. CONCLUSIONS: Decreased expression of hMYCN protein is achievable with the use of AS oligonucleotide treatment, even in the presence of hMYCN oncogene amplification. Antisense strategies targeting the hMYCN oncogene in vivo decrease mouse neuroblastoma tumorigenesis. Investigation of their clinical effect in children with neuroblastoma is warranted.


Subject(s)
Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Nuclear Proteins/pharmacology , Oligonucleotides, Antisense/pharmacology , Oncogene Proteins/pharmacology , Animals , Disease Models, Animal , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Nuclear Proteins/administration & dosage , Nuclear Proteins/genetics , Oligonucleotides, Antisense/administration & dosage , Oncogene Proteins/administration & dosage , Oncogene Proteins/genetics , Transfection
11.
J Biochem Biophys Methods ; 52(2): 145-9, 2002 Jul 31.
Article in English | MEDLINE | ID: mdl-12204418

ABSTRACT

Although real-time PCR is a rapid, quantitative method for the analysis of gene and RNA levels, the presence of inhibitors in samples is an obstacle to its successful use. We have found that genomic DNA isolated from mouse tail tips using a standard proteinase K digestion method caused marked inhibition of real-time PCR. Inhibition was specific for mouse tail DNA since genomic DNA isolated from other tissue sources using the same methodology was readily amplified. We have therefore developed a nonproteinase K DNA isolation method involving the use of Chelex 100 resin. This method produces mouse tail DNA that is free of real-time PCR inhibitors.


Subject(s)
DNA/genetics , DNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Endopeptidase K/pharmacology , Mice , Mice, Transgenic , Tail/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...