Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Org Chem ; 82(11): 5881-5889, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28493695

ABSTRACT

A method for the preparation of 1-(N-ribofuranosyl)-6-imino-1,6-dihydropyrimidin-4-amines 3 or 4-(N-ribofuranosyl)-6-aminopyrimidines 4 via glycosylation of 4-aminopyrimidines 2 or 5 is described. Silylated 4-aminopyrimidines 2 or 5 upon ribosylation with 1 provide products 3. When intermediates 3 contain a strongly electron-withdrawing group, such as C(4)-Cl or C(5)-NO2, they rearrange to products 4 in the presence of aqueous ammonia. A mechanism is proposed that involves a ring-opening/ring-closing (Dimroth) rearrangement.

2.
Nat Commun ; 12(1): 7299, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911927

ABSTRACT

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA Splicing , Small Molecule Libraries/administration & dosage , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Humans , Huntington Disease/metabolism , Mice , RNA Splicing/drug effects , RNA Stability/drug effects , Trinucleotide Repeat Expansion/drug effects
3.
Oncogene ; 38(10): 1702-1716, 2019 03.
Article in English | MEDLINE | ID: mdl-30348991

ABSTRACT

Medulloblastoma (MB) is the most frequent malignant pediatric brain tumor, representing 20% of newly diagnosed childhood central nervous system malignancies. Although advances in multimodal therapy yielded a 5-year survivorship of 80%, MB still accounts for the leading cause of childhood cancer mortality. In this work, we describe the epigenetic regulator BMI1 as a novel therapeutic target for the treatment of recurrent human Group 3 MB, a childhood brain tumor for which there is virtually no treatment option beyond palliation. Current clinical trials for recurrent MB patients based on genomic profiles of primary, treatment-naive tumors will provide limited clinical benefit since recurrent metastatic MBs are highly genetically divergent from their primary tumor. Using a small molecule inhibitor against BMI1, PTC-028, we were able to demonstrate complete ablation of self-renewal of MB stem cells in vitro. When administered to mice xenografted with patient tumors, we observed significant reduction in tumor burden in both local and metastatic compartments and subsequent increased survival, without neurotoxicity. Strikingly, serial in vivo re-transplantation assays demonstrated a marked reduction in tumor initiation ability of recurrent MB cells upon re-transplantation of PTC-028-treated cells into secondary recipient mouse brains. As Group 3 MB is often metastatic and uniformly fatal at recurrence, with no current or planned trials of targeted therapy, an efficacious targeted agent would be rapidly transitioned to clinical trials.


Subject(s)
Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Polycomb Repressive Complex 1/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Child , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Small Molecule Libraries/pharmacology , Treatment Outcome , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 17(1): 39-49, 2018 01.
Article in English | MEDLINE | ID: mdl-29158468

ABSTRACT

BMI-1, also known as a stem cell factor, is frequently upregulated in several malignancies. Elevated expression of BMI-1 correlates with poor prognosis and is therefore considered a viable therapeutic target in a number of malignancies including ovarian cancer. Realizing the immense pathologic significance of BMI-1, small-molecule inhibitors against BMI-1 are recently being developed. In this study, we functionally characterize PTC-028, an orally bioavailable compound that decreases BMI-1 levels by posttranslational modification. We report that PTC-028 treatment selectively inhibits cancer cells in clonal growth and viability assays, whereas normal cells remain unaffected. Mechanistically, hyperphosphorylation-mediated depletion of cellular BMI-1 by PTC-028 coupled with a concurrent temporal decrease in ATP and a compromised mitochondrial redox balance potentiates caspase-dependent apoptosis. In vivo, orally administered PTC-028, as a single agent, exhibits significant antitumor activity comparable with the standard cisplatin/paclitaxel therapy in an orthotopic mouse model of ovarian cancer. Thus, PTC-028 has the potential to be used as an effective therapeutic agent in patients with epithelial ovarian cancer, where treatment options are limited. Mol Cancer Ther; 17(1); 39-49. ©2017 AACR.


Subject(s)
Benzimidazoles/pharmacology , Carcinoma, Ovarian Epithelial/drug therapy , Polycomb Repressive Complex 1/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Xenograft Model Antitumor Assays
5.
Org Lett ; 8(12): 2611-4, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16737326

ABSTRACT

Torquoselective pericyclic ring closures of 1-azatrienes that contain acyclic chirality at the C-terminus are described herein. [reaction: see text]


Subject(s)
Aza Compounds/chemistry , Aza Compounds/chemical synthesis , Catalysis , Crystallography, X-Ray , Electrochemistry/methods , Molecular Conformation , Molecular Structure
6.
Org Lett ; 8(2): 191-3, 2006 Jan 19.
Article in English | MEDLINE | ID: mdl-16408872

ABSTRACT

[reaction: see text] A Lewis acid-catalyzed formal cycloaddition of alpha,beta-unsaturated aldehydes with 6-methyl-4-hydroxy-2-pyrone, 1,3-diketones, and vinylogous silyl esters is described here.


Subject(s)
Aldehydes/chemistry , Ketones/chemistry , Pyrans/chemical synthesis , Pyrones/chemistry , Vinyl Compounds/chemistry , Catalysis , Cyclization , Esters , Indicators and Reagents , Molecular Structure , Pyrans/chemistry , Stereoisomerism
7.
Clin Cancer Res ; 22(24): 6176-6191, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27307599

ABSTRACT

PURPOSE: Current prostate cancer management calls for identifying novel and more effective therapies. Self-renewing tumor-initiating cells (TICs) hold intrinsic therapy resistance and account for tumor relapse and progression. As BMI-1 regulates stem cell self-renewal, impairing BMI-1 function for TIC-tailored therapies appears to be a promising approach. EXPERIMENTAL DESIGN: We have previously developed a combined immunophenotypic and time-of-adherence assay to identify CD49bhiCD29hiCD44hi cells as human prostate TICs. We utilized this assay with patient-derived prostate cancer cells and xenograft models to characterize the effects of pharmacologic inhibitors of BMI-1. RESULTS: We demonstrate that in cell lines and patient-derived TICs, BMI-1 expression is upregulated and associated with stem cell-like traits. From a screened library, we identified a number of post-transcriptional small molecules that target BMI-1 in prostate TICs. Pharmacologic inhibition of BMI-1 in patient-derived cells significantly decreased colony formation in vitro and attenuated tumor initiation in vivo, thereby functionally diminishing the frequency of TICs, particularly in cells resistant to proliferation- and androgen receptor-directed therapies, without toxic effects on normal tissues. CONCLUSIONS: Our data offer a paradigm for targeting TICs and support the development of BMI-1-targeting therapy for a more effective prostate cancer treatment. Clin Cancer Res; 22(24); 6176-91. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Self Renewal/drug effects , Cell Survival/drug effects , Neoplastic Stem Cells/drug effects , Polycomb Repressive Complex 1/metabolism , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays/methods
8.
Sci Transl Med ; 8(350): 350ra104, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27488898

ABSTRACT

Lung cancer is the most common cause of cancer deaths. The expression of the transcription factor C/EBPα (CCAAT/enhancer binding protein α) is frequently lost in non-small cell lung cancer, but the mechanisms by which C/EBPα suppresses tumor formation are not fully understood. In addition, no pharmacological therapy is available to specifically target C/EBPα expression. We discovered a subset of pulmonary adenocarcinoma patients in whom negative/low C/EBPα expression and positive expression of the oncogenic protein BMI1 (B lymphoma Mo-MLV insertion region 1 homolog) have prognostic value. We also generated a lung-specific mouse model of C/EBPα deletion that develops lung adenocarcinomas, which are prevented by Bmi1 haploinsufficiency. BMI1 activity is required for both tumor initiation and maintenance in the C/EBPα-null background, and pharmacological inhibition of BMI1 exhibits antitumor effects in both murine and human adenocarcinoma lines. Overall, we show that C/EBPα is a tumor suppressor in lung cancer and that BMI1 is required for the oncogenic process downstream of C/EBPα loss. Therefore, anti-BMI1 pharmacological inhibition may offer a therapeutic benefit for lung cancer patients with low expression of C/EBPα and high BMI1.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/therapy , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Knockout , Mutation/genetics , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics
9.
Nat Med ; 20(1): 29-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292392

ABSTRACT

Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key 'stemness' property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1-related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Heterocyclic Compounds, 2-Ring/pharmacology , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Thiazoles/pharmacology , Animals , Blotting, Western , Bromodeoxyuridine , Cell Line, Tumor , Flow Cytometry , Genetic Vectors/genetics , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Luciferases , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Thiazoles/therapeutic use
10.
J Org Chem ; 70(11): 4248-56, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15903297

ABSTRACT

A detailed account on chiral secondary amine salt promoted enantioselective intramolecular formal aza-[3 + 3] cycloadditions is described here for the first time. The dependence of enantioselectivity on the structural feature of these chiral amines is thoroughly investigated. This study also reveals a very interesting reversal of the stereochemistry in the respective cycloadducts obtained using C(1)- and C(2)-symmetric amine salts. In addition, the influence of solvents, counteranions, and temperatures on the enantioselectivity is described, and a unified mechanistic model based on experimental results as well as semiempirical calculations is proposed.


Subject(s)
Amines/chemistry , Heterocyclic Compounds/chemical synthesis , Models, Chemical , Catalysis , Crystallography, X-Ray , Cyclization , Molecular Conformation , Molecular Structure , Salts/chemistry , Stereoisomerism
11.
Org Biomol Chem ; 3(11): 2140-4, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15917902

ABSTRACT

Total syntheses of indoloquinolizidine alkaloid (+/-)-, R-(+)-, and S-(-)-deplancheine are described here. The synthesis features an enantioselective intramolecular formal aza-[3 + 3] cycloaddition for the construction of the quinolizidine CD-ring. This application serves to introduce a new synthetic strategy for the synthesis of indoloquinolizidine alkaloids.


Subject(s)
Indole Alkaloids/chemical synthesis , Indole Alkaloids/chemistry , Magnetic Resonance Spectroscopy , Stereoisomerism
12.
J Org Chem ; 69(20): 6732-8, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15387597

ABSTRACT

A detailed account regarding formal aza-[3 + 3] cycloaddition reactions of tetronamides with alpha,beta-unsaturated iminium salts is described here. This investigation uncovers regioisomeric cycloadducts that were not found in previous studies involving this formal cycloaddition and an unexpected rearrangement that led to pyridines and dihydropyridines. Both stereochemical and regiochemical issues raised in this study provide further mechanistic insights into this cycloaddition. With careful control of reaction temperatures, the desired formal cycloadducts are obtained. Ensuing transformation of these cycloadducts into functionalized piperidines establishes the concept of employing tetronamides as latent acyclic vinylogous amides for the formal aza-[3 + 3] cycloaddition.


Subject(s)
Amides/chemistry , Piperidines/chemical synthesis , Imines/chemistry , Models, Chemical , Molecular Structure , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL