Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(36): e2302342120, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37639589

ABSTRACT

Inhibition of overexpressed enzymes is among the most promising approaches for targeted cancer treatment. However, many cancer-expressed enzymes are "nonlethal," in that the inhibition of the enzymes' activity is insufficient to kill cancer cells. Conventional antibody-based therapeutics can mediate efficient treatment by targeting extracellular nonlethal targets but can hardly target intracellular enzymes. Herein, we report a cancer targeting and treatment strategy to utilize intracellular nonlethal enzymes through a combination of selective cancer stem-like cell (CSC) labeling and Click chemistry-mediated drug delivery. A de novo designed compound, AAMCHO [N-(3,4,6-triacetyl- N-azidoacetylmannosamine)-cis-2-ethyl-3-formylacrylamideglycoside], selectively labeled cancer CSCs in vitro and in vivo through enzymatic oxidation by intracellular aldehyde dehydrogenase 1A1. Notably, azide labeling is more efficient in identifying tumorigenic cell populations than endogenous markers such as CD44. A dibenzocyclooctyne (DBCO)-toxin conjugate, DBCO-MMAE (Monomethylauristatin E), could next target the labeled CSCs in vivo via bioorthogonal Click reaction to achieve excellent anticancer efficacy against a series of tumor models, including orthotopic xenograft, drug-resistant tumor, and lung metastasis with low toxicity. A 5/7 complete remission was observed after single-cycle treatment of an advanced triple-negative breast cancer xenograft (~500 mm3).


Subject(s)
Aldehyde Dehydrogenase , Antibodies , Humans , Azides , Carcinogenesis , Click Chemistry , Aldehyde Dehydrogenase 1 Family , Retinal Dehydrogenase
2.
ACS Med Chem Lett ; 12(6): 935-940, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34141072

ABSTRACT

Myotonic dystrophy type 1 originates from d(CTG·CAG) repeats that undergo aberrant expansion during normal processing because the d(CTG) repeat forms stable hairpin structures. Bidirectional transcription of d(CTG·CAG) yields two RNA transcripts that undergo repeat-associated non-ATG (RAN) translation to form homopolymeric proteins. Thus, both the r(CUG) transcript and the r(CAG) transcript are known to be toxic. We report a pairwise fragment-based, target-guided approach to screen for proximity-induced click dimers formed on the nucleic acid template. This screen uses an azide/alkyne clickable fragment library of nucleic acid-binding ligands incubated in parallel, pairwise reactions as an alternative to our previously reported one-pot screening method. MALDI-TOF mass spectroscopy was used to detect template assisted click products. Hit compounds inhibited the in vitro transcription of d(CTG·CAG)90 bidirectionally with IC50 values in the low micromolar range. This approach may be broadly applicable to other trinucleotide repeat diseases and in targeting other disease-associated nucleic acid sequences.

3.
Biochemistry ; 59(37): 3463-3472, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32856901

ABSTRACT

There are few methods available for the rapid discovery of multitarget drugs. Herein, we describe the template-assisted, target-guided discovery of small molecules that recognize d(CTG) in the expanded d(CTG·CAG) sequence and its r(CUG) transcript that cause myotonic dystrophy type 1. A positive cross-selection was performed using a small library of 30 monomeric alkyne- and azide-containing ligands capable of producing >5000 possible di- and trimeric click products. The monomers were incubated with d(CTG)16 or r(CUG)16 under physiological conditions, and both sequences showed selectivity in the proximity-accelerated azide-alkyne [3+2] cycloaddition click reaction. The limited number of click products formed in both selections and the even smaller number of common products suggests that this method is a useful tool for the discovery of single-target and multitarget lead therapeutic agents.


Subject(s)
DNA/antagonists & inhibitors , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , RNA/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cells, Cultured , DNA/genetics , DNA/metabolism , Humans , Myotonic Dystrophy/pathology , RNA/genetics , RNA/metabolism , Trinucleotide Repeat Expansion/genetics
4.
Proc Natl Acad Sci U S A ; 116(18): 8709-8714, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30975744

ABSTRACT

Developing highly active, multivalent ligands as therapeutic agents is challenging because of delivery issues, limited cell permeability, and toxicity. Here, we report intrinsically cell-penetrating multivalent ligands that target the trinucleotide repeat DNA and RNA in myotonic dystrophy type 1 (DM1), interrupting the disease progression in two ways. The oligomeric ligands are designed based on the repetitive structure of the target with recognition moieties alternating with bisamidinium groove binders to provide an amphiphilic and polycationic structure, mimicking cell-penetrating peptides. Multiple biological studies suggested the success of our multivalency strategy. The designed oligomers maintained cell permeability and exhibited no apparent toxicity both in cells and in mice at working concentrations. Furthermore, the oligomers showed important activities in DM1 cells and in a DM1 liver mouse model, reducing or eliminating prominent DM1 features. Phenotypic recovery of the climbing defect in adult DM1 Drosophila was also observed. This design strategy should be applicable to other repeat expansion diseases and more generally to DNA/RNA-targeted therapeutics.


Subject(s)
Myotonic Dystrophy/drug therapy , RNA-Binding Proteins/metabolism , Trinucleotide Repeats , Animals , DNA , DNA-Binding Proteins , Drosophila melanogaster , HeLa Cells , Humans , Ligands , Liver/metabolism , Mice , Myoblasts/physiology , Myotonic Dystrophy/genetics , RNA Recognition Motif Proteins , RNA-Binding Proteins/chemistry
5.
ChemMedChem ; 11(13): 1428-35, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27245480

ABSTRACT

Myotonic dystrophy is the most common form of adult-onset muscular dystrophy, originating in a CTG repeat expansion in the DMPK gene. The expanded CUG transcript sequesters MBNL1, a key regulator of alternative splicing, leading to the misregulation of numerous pre-mRNAs. We report an RNA-targeted agent as a possible lead compound for the treatment of myotonic dystrophy type 1 (DM1) that reveals both the promise and challenges for this type of small-molecule approach. The agent is a potent inhibitor of the MBNL1-rCUG complex with an inhibition constant (Ki ) of 25±8 nm, and is also relatively nontoxic to HeLa cells, able to dissolve nuclear foci, and correct the insulin receptor splicing defect in DM1 model cells. Moreover, treatment with this compound improves two separate disease phenotypes in a Drosophila model of DM1: adult external eye degeneration and larval crawling defect. However, the compound has a relatively low maximum tolerated dose in mice, and its cell uptake may be limited, providing insight into directions for future development.


Subject(s)
Amidines/therapeutic use , Myotonic Dystrophy/drug therapy , RNA-Binding Proteins/antagonists & inhibitors , Triazines/therapeutic use , Trinucleotide Repeat Expansion , Amidines/chemical synthesis , Amidines/pharmacology , Animals , Animals, Genetically Modified , Carbocyanines/chemistry , Click Chemistry , Cycloaddition Reaction , Drosophila , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Ligands , Mice , RNA/antagonists & inhibitors , RNA Splicing/drug effects , Receptor, Insulin/genetics , Triazines/chemical synthesis , Triazines/pharmacology
6.
J Am Chem Soc ; 137(44): 14180-9, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26473464

ABSTRACT

Single-agent, single-target therapeutic approaches are often limited by a complex disease pathobiology. We report rationally designed, multi-target agents for myotonic dystrophy type 1 (DM1). DM1 originates in an abnormal expansion of CTG repeats (CTG(exp)) in the DMPK gene. The resultant expanded CUG transcript (CUG(exp)) identified as a toxic agent sequesters important proteins, such as muscleblind-like proteins (MBNL), undergoes repeat-associated non-ATG (RAN) translation, and potentially causes microRNA dysregulation. We report rationally designed small molecules that target the DM1 pathobiology in vitro in three distinct ways by acting simultaneously as transcription inhibitors, by inhibiting aberrant protein binding to the toxic RNA, and by acting as RNase mimics to degrade the toxic RNA. In vitro, the agents are shown to (1) bind CTG(exp) and inhibit formation of the CUG(exp) transcript, (2) bind CUG(exp) and inhibit sequestration of MBNL1, and (3) cleave CUG(exp) in an RNase-like manner. The most potent compounds are capable of reducing the levels of CUG(exp) in DM1 model cells, and one reverses two separate CUG(exp)-induced phenotypes in a DM1 Drosophila model.


Subject(s)
DNA/metabolism , Drug Design , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , RNA/metabolism , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Animals , DNA/genetics , Disease Models, Animal , Drosophila , HeLa Cells , Humans , Myotonic Dystrophy/pathology , RNA/biosynthesis , RNA/genetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Trinucleotide Repeat Expansion/genetics
7.
J Am Chem Soc ; 136(17): 6355-61, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24702247

ABSTRACT

A working hypothesis for the pathogenesis of myotonic dystrophy type 1 (DM1) involves the aberrant sequestration of an alternative splicing regulator, MBNL1, by expanded CUG repeats, r(CUG)(exp). It has been suggested that a reversal of the myotonia and potentially other symptoms of the DM1 disease can be achieved by inhibiting the toxic MBNL1-r(CUG)(exp) interaction. Using rational design, we discovered an RNA-groove binding inhibitor (ligand 3) that contains two triaminotriazine units connected by a bisamidinium linker. Ligand 3 binds r(CUG)12 with a low micromolar affinity (K(d) = 8 ± 2 µM) and disrupts the MBNL1-r(CUG)12 interaction in vitro (K(i) = 8 ± 2 µM). In addition, ligand 3 is cell and nucleus permeable, exhibits negligible toxicity to mammalian cells, dissolves MBNL1-r(CUG)(exp) ribonuclear foci, and restores misregulated splicing of IR and cTNT in a DM1 cell culture model. Importantly, suppression of r(CUG)(exp) RNA-induced toxicity in a DM1 Drosophila model was observed after treatment with ligand 3. These results suggest ligand 3 as a lead for the treatment of DM1.


Subject(s)
DNA-Binding Proteins/metabolism , Imidazoles/chemistry , Imidazoles/pharmacology , Myotonic Dystrophy/genetics , RNA-Binding Proteins/metabolism , RNA/genetics , Trinucleotide Repeat Expansion/drug effects , Alternative Splicing/drug effects , Animals , Base Sequence , DNA-Binding Proteins/antagonists & inhibitors , Drosophila , Drug Discovery , HeLa Cells , Humans , Mice, Inbred C57BL , Models, Molecular , Molecular Targeted Therapy , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/metabolism , Nucleic Acid Conformation/drug effects , RNA/antagonists & inhibitors , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/antagonists & inhibitors
8.
J Med Chem ; 56(23): 9471-9481, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24188018

ABSTRACT

An expanded CUG repeat transcript (CUG(exp)) is the causative agent of myotonic dystrophy type 1 (DM1) by sequestering muscleblind-like 1 protein (MBNL1), a regulator of alternative splicing. On the basis of a ligand (1) that was previously reported to be active in an in vitro assay, we present the synthesis of a small library containing 10 dimeric ligands (4-13) that differ in length, composition, and attachment point of the linking chain. The oligoamino linkers gave a greater gain in affinity for CUG RNA and were more effective when compared to oligoether linkers. The most potent in vitro ligand (9) was shown to be aqueous-soluble and both cell- and nucleus-permeable, displaying almost complete dispersion of MBNL1 ribonuclear foci in a DM1 cell model. Direct evidence for the bioactivity of 9 was observed in its ability to disperse ribonuclear foci in individual live DM1 model cells using time-lapse confocal fluorescence microscopy.


Subject(s)
Myotonic Dystrophy/genetics , Polyamines/chemical synthesis , Trinucleotide Repeats , Humans , Ligands , Microscopy, Confocal , Polyamines/pharmacology , RNA/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...