Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 519
Filter
1.
Commun Biol ; 7(1): 963, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122837

ABSTRACT

Limiting cellular plasticity is of key importance for the therapeutic targeting of metastatic breast cancer (MBC). Fibroblast growth receptor (FGFR) is a critical molecule in cellular plasticity and potent inhibitors of FGFR enzymatic activity have been developed, but kinase independent functions for this receptor also contribute to MBC progression. Herein, we evaluated several FGFR inhibitors and find that while FGFR-targeted kinase inhibitors are effective at blocking ligand-induced cell growth, dormant cells persist eventually giving rise to MBC progression. To more broadly target FGFR and cellular plasticity, we examined the FGFR1 proximal promoter, and found several sequences with potential to form G-quadruplex secondary structures. Circular dichroism was used to verify formation of G-quadruplex in the FGFR1 proximal promoter. Importantly, use of the clinical G-quadruplex-stabilizing compound, CX-5461, stabilized the FGFR1 G-quadruplex structures, blocked the transcriptional activity of the FGFR1 proximal promoter, decreased FGFR1 expression, and resulted in potent inhibition of pulmonary tumor formation. Overall, our findings suggest G-quadruplex-targeted compounds could be a potential therapeutic strategy to limit the cellular plasticity of FGFR1 overexpressing MBC.


Subject(s)
Breast Neoplasms , G-Quadruplexes , Promoter Regions, Genetic , Receptor, Fibroblast Growth Factor, Type 1 , G-Quadruplexes/drug effects , Humans , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Female , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Animals , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis
2.
Molecules ; 29(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125057

ABSTRACT

G-quadruplex (G4) sequences, which can fold into higher-order G4 structures, are abundant in the human genome and are over-represented in the promoter regions of many genes involved in human cancer initiation, progression, and metastasis. They are plausible targets for G4-binding small molecules, which would, in the case of promoter G4s, result in the transcriptional downregulation of these genes. However, structural information is currently available on only a very small number of G4s and their ligand complexes. This limitation, coupled with the currently restricted information on the G4-containing genes involved in most complex human cancers, has led to the development of a phenotypic-led approach to G4 ligand drug discovery. This approach was illustrated by the discovery of several generations of tri- and tetra-substituted naphthalene diimide (ND) ligands that were found to show potent growth inhibition in pancreatic cancer cell lines and are active in in vivo models for this hard-to-treat disease. The cycles of discovery have culminated in a highly potent tetra-substituted ND derivative, QN-302, which is currently being evaluated in a Phase 1 clinical trial. The major genes whose expression has been down-regulated by QN-302 are presented here: all contain G4 propensity and have been found to be up-regulated in human pancreatic cancer. Some of these genes are also upregulated in other human cancers, supporting the hypothesis that QN-302 is a pan-G4 drug of potential utility beyond pancreatic cancer.


Subject(s)
Antineoplastic Agents , Drug Discovery , G-Quadruplexes , G-Quadruplexes/drug effects , Humans , Drug Discovery/methods , Ligands , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Phenotype , Cell Line, Tumor , Naphthalenes/pharmacology , Naphthalenes/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Animals , Imides/chemistry , Imides/pharmacology , Promoter Regions, Genetic
3.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3421-3431, 2024 Jul.
Article in Chinese | MEDLINE | ID: mdl-39041114

ABSTRACT

DNA G-quadruplex(G4) is a guanine-rich single-stranded DNA sequence that spontaneously folds into a spherical four-stranded DNA secondary structure in oncogene promoter sequences and telomeres. G4s are highly associated with the occurrence and development of cancer and have emerged as promising anticancer targets. Natural products have long been important sources of anticancer drug development. In recent years, significant progress has been made in the discovery of natural drugs targeting DNA G4s, with many DNA G4s have been confirmed as promising targets of natural products, including MYC-G4, KRAS-G4, PDGFR-ß-G4, BCL-2-G4, VEGF-G4, and telomeric G4. This review summarizes the research progress in discovering natural small molecules that target DNA G4s and their binding mechanisms. It also discusses the opportunities of and challenges in developing drugs targeting DNA G4s. This review will serve as a valuable reference for the research on natural products, particularly in the development of novel antitumor medications.


Subject(s)
Biological Products , G-Quadruplexes , G-Quadruplexes/drug effects , Biological Products/chemistry , Biological Products/pharmacology , Humans , Animals , DNA/chemistry , DNA/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
4.
J Med Chem ; 67(15): 13363-13382, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38987863

ABSTRACT

Human telomerase reverse transcriptase (hTERT) may have noncanonical functions in transcriptional regulation and metabolic reprogramming in cancer cells, but it is a challenging target. We thus developed small-molecule ligands targeting hTERT promoter G-quadruplex DNA structures (hTERT G4) to downregulate hTERT expression. Ligand 5 showed high affinity toward hTERT G4 (Kd = 1.1 µM) and potent activity against triple-negative breast cancer cells (MDA-MB-231, IC50 = 1 µM). In cell-based assays, 5 not only exerts markedly inhibitory activity on classical telomere functions including decreased telomerase activity, shortened telomere length, and cellular senescence but also induces DNA damage, acute cellular senescence, and apoptosis. This study reveals that hTERT G4-targeting ligand may cause mitochondrial dysfunction, disrupt iron metabolism and activate ferroptosis in cancer cells. The in vivo antitumor efficacy of 5 was also evaluated in an MDA-MB-231 xenograft mouse model and approximately 78.7% tumor weight reduction was achieved. No observable toxicity against the major organs was observed.


Subject(s)
Antineoplastic Agents , Down-Regulation , G-Quadruplexes , Promoter Regions, Genetic , Telomerase , Triple Negative Breast Neoplasms , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Humans , G-Quadruplexes/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Ligands , Female , Down-Regulation/drug effects , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Mice, Nude , Cellular Senescence/drug effects , Mice, Inbred BALB C
5.
Bioorg Med Chem Lett ; 111: 129903, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39053704

ABSTRACT

Nitrobenzoxadiazole (NBD)-incorporated naphthalene diimide derivatives were designed and synthesized as candidates of antitumor agents with cytotoxicity against human pancreatic cancer cell MIA PaCa-2. Among these, compounds 1NND and 3NND exhibited fluorescent "turn-off" property toward human telomeric G-quadruplex (G4), which allows the direct measurement of dissociation constant (Kd) of ligands against G4 by fluorescence titration method. Notably, the compound 1NND not only exhibited great cytotoxic activity against MIA PaCa-2 with a half maximal inhibitory concentration (IC50) of 77.9 nM, but also exhibited high affinity against G4 with Kd of 1.72 µM. Furthermore, the target binding properties were investigated by circular dichroism (CD) spectra and further studied by molecular docking methods.


Subject(s)
Antineoplastic Agents , Drug Design , G-Quadruplexes , Imides , Naphthalenes , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/pharmacology , G-Quadruplexes/drug effects , Imides/chemistry , Imides/pharmacology , Imides/chemical synthesis , Ligands , Molecular Docking Simulation , Molecular Structure , Naphthalenes/chemistry , Naphthalenes/pharmacology , Naphthalenes/chemical synthesis , Structure-Activity Relationship
6.
Eur J Med Chem ; 276: 116641, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38971047

ABSTRACT

Chagas disease is caused by the parasite Trypanosoma cruzi and affects over 7 million people worldwide. The two actual treatments, Benznidazole (Bzn) and Nifurtimox, cause serious side effects due to their high toxicity leading to treatment abandonment by the patients. In this work, we propose DNA G-quadruplexes (G4) as potential therapeutic targets for this infectious disease. We have found 174 PQS per 100,000 nucleotides in the genome of T. cruzi and confirmed G4 formation of three frequent motifs. We synthesized a family of 14 quadruplex ligands based in the dithienylethene (DTE) scaffold and demonstrated their binding to these identified G4 sequences. Several DTE derivatives exhibited micromolar activity against epimastigotes of four different strains of T. cruzi, in the same concentration range as Bzn. Compounds L3 and L4 presented remarkable activity against trypomastigotes, the active form in blood, of T. cruzi SOL strain (IC50 = 1.5-3.3 µM, SI = 25-40.9), being around 40 times more active than Bzn and displaying much better selectivity indexes.


Subject(s)
Chagas Disease , G-Quadruplexes , Trypanocidal Agents , Trypanosoma cruzi , Trypanosoma cruzi/drug effects , G-Quadruplexes/drug effects , Ligands , Chagas Disease/drug therapy , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/chemical synthesis , Humans , Molecular Structure , Structure-Activity Relationship , Dose-Response Relationship, Drug , Parasitic Sensitivity Tests , Antiparasitic Agents/pharmacology , Antiparasitic Agents/chemistry , Antiparasitic Agents/chemical synthesis
7.
J Enzyme Inhib Med Chem ; 39(1): 2366236, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38905127

ABSTRACT

A novel class of compounds designed to hit two anti-tumour targets, G-quadruplex structures and human carbonic anhydrases (hCAs) IX and XII is proposed. The induction/stabilisation of G-quadruplex structures by small molecules has emerged as an anticancer strategy, disrupting telomere maintenance and reducing oncogene expression. hCAs IX and XII are well-established anti-tumour targets, upregulated in many hypoxic tumours and contributing to metastasis. The ligands reported feature a berberine G-quadruplex stabiliser scaffold connected to a moiety inhibiting hCAs IX and XII. In vitro experiments showed that our compounds selectively stabilise G-quadruplex structures and inhibit hCAs IX and XII. The crystal structure of a telomeric G-quadruplex in complex with one of these ligands was obtained, shedding light on the ligand/target interaction mode. The most promising ligands showed significant cytotoxicity against CA IX-positive HeLa cancer cells in hypoxia, and the ability to stabilise G-quadruplexes within tumour cells.


Subject(s)
Antineoplastic Agents , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors , Carbonic Anhydrases , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , G-Quadruplexes , Humans , G-Quadruplexes/drug effects , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Molecular Structure , Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrases/metabolism , Cell Proliferation/drug effects , Ligands , HeLa Cells , Antigens, Neoplasm/metabolism , Models, Molecular
8.
Bioorg Med Chem Lett ; 109: 129855, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38908766

ABSTRACT

The role of G-quadruplex (G4) in cellular processes can be investigated by the covalent modification of G4-DNA using alkylating reagents. Controllable alkylating reagents activated by external stimuli can react elegantly and selectively. Herein, we report a chemical activation system that can significantly boost the reaction rate of methylamine-protected vinyl-quinazolinone (VQ) derivative for the alkylation of G4-DNA. The two screened activators can transform low-reactive VQ-NHR' to highly reactive intermediates following the Michael addition mechanism. This approach expands the toolbox of activable G4 alkylating reagents.


Subject(s)
G-Quadruplexes , Methylamines , Quinazolinones , Alkylation , G-Quadruplexes/drug effects , Methylamines/chemistry , Methylamines/pharmacology , Methylamines/chemical synthesis , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemical synthesis , Humans , Molecular Structure , DNA/chemistry , Vinyl Compounds/chemistry , Vinyl Compounds/pharmacology
9.
J Med Chem ; 67(13): 10643-10654, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38924701

ABSTRACT

Several G-quadruplex nucleic acid (G4s) ligands have been developed seeking target selectivity in the past decade. Naphthalene diimide (NDI)-based compounds are particularly promising due to their biological activity and red-fluorescence emission. Previously, we demonstrated the existence of G4s in the promoter region of parasite genomes, assessing the effectiveness of NDI-derivatives against them. Here, we explored the biological activity of a small library of G4-DNA ligands, exploiting the NDI pharmacophore, against both Trypanosoma brucei and Leishmania major parasites. Biophysical and biological assays were conducted. Among the various families analyzed, core-extended NDIs exhibited the most promising results concerning the selectivity and antiparasitic effects. NDI 16 emerged as the most potent, with an IC50 of 0.011 nM against T. brucei and remarkable selectivity vs MRC-5 cells (3454-fold). Fascinating, 16 is 480-fold more potent than the standard drug pentamidine (IC50 = 5.3 nM). Cellular uptake and parasite localization were verified by exploiting core-extended NDI red-fluorescent emission.


Subject(s)
G-Quadruplexes , Imides , Leishmania major , Naphthalenes , Trypanocidal Agents , Trypanosoma brucei brucei , G-Quadruplexes/drug effects , Structure-Activity Relationship , Naphthalenes/pharmacology , Naphthalenes/chemistry , Imides/chemistry , Imides/pharmacology , Ligands , Trypanosoma brucei brucei/drug effects , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/chemical synthesis , Humans , Leishmania major/drug effects , Cell Line
10.
Comput Biol Med ; 177: 108683, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38838555

ABSTRACT

G-Quadruplex DNA (GQ-DNA) is one of the most important non-canonical nucleic acid structures. GQ-DNA forming sequences are present in different crucial genomic regions and are abundant in promoter regions of several oncogenes. Therefore, GQ-DNA is an important target for anticancer drugs and hence binding interactions between GQ-DNA and small molecule ligands are of great importance. Since GQ-DNA is a highly polymorphic structure, it is important to identify ligand molecules which preferentially target a particular quadruplex sequence. In this present study, we have used a FDA approved drug called imatinib mesylate (ligand) which is a selective tyrosine kinase inhibitor, successfully used for the treatment of chronic myelogenous leukaemia, gastrointestinal stromal tumours. Different spectroscopic techniques as well as molecular docking investigations and molecular simulations have been used to explore the interaction between imatinib mesylate with VEGF GQ DNA structures along with duplex DNA, C-Myc, H-Telo GQ DNA. We found that imatinib mesylate shows preferential interaction towards VEGF GQ DNA compared to C-Myc, H-Telo GQ and duplex DNA. Imatinib mesylate seems to be an efficient ligand for VEGF GQ DNA, suggesting that it might be used to regulate the expression of genes in cancerous cells.


Subject(s)
Antineoplastic Agents , G-Quadruplexes , Imatinib Mesylate , Molecular Docking Simulation , Vascular Endothelial Growth Factor A , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/chemistry , Imatinib Mesylate/pharmacology , G-Quadruplexes/drug effects , Humans , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , DNA/chemistry , DNA/metabolism
11.
Nanoscale ; 16(26): 12502-12509, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38873939

ABSTRACT

Targeted protein degradation through PROteolysis TArgeting Chimeras (PROTACs) is a relatively new modality in cellular interventions. The minimum requirement for PROTACs to function is forming a tertiary complex of the protein of interest (POI), E3 ligase, and the molecular glue PROTAC. Here, we propose a new approach to modulate the nano-environment interactome of a non-protein target through a plausible quaternary complex of interactome-biomolecule of interest (BOI)-PROTAC and E3 ligase. We report nucleic acid-targeting PROTAC (NA-TAC) molecules by conjugating DNA-binding and E3 ligase ligands. We demonstrate that NA-TACs can target the G-quadruplex DNA and induce elevated DNA damage and cytotoxicity compared to the conventional G-quadruplex binding ligands. Our new class of NA-TACs lays the foundation for small molecule-based non-protein targeting PROTACs for interactome and nanoenvironment mapping and nucleic acid-targeted precision medicines.


Subject(s)
Antineoplastic Agents , G-Quadruplexes , Proteolysis , Ubiquitin-Protein Ligases , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Proteolysis/drug effects , Ubiquitin-Protein Ligases/metabolism , G-Quadruplexes/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Ligands , Nucleic Acids/chemistry , Nucleic Acids/metabolism , DNA/chemistry , DNA/metabolism , Proteolysis Targeting Chimera
12.
Nucleic Acids Res ; 52(12): 6748-6762, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38828773

ABSTRACT

Noncanonical nucleic acid structures, particularly G-quadruplexes, have garnered significant attention as potential therapeutic targets in cancer treatment. Here, the recognition of G-quadruplex DNA by peptides derived from the Rap1 protein is explored, with the aim of developing novel peptide-based G-quadruplex ligands with enhanced selectivity and anticancer activity. Biophysical techniques were employed to assess the interaction of a peptide derived from the G-quadruplex-binding domain of the protein with various biologically relevant G-quadruplex structures. Through alanine scanning mutagenesis, key amino acids crucial for G-quadruplex recognition were identified, leading to the discovery of two peptides with improved G-quadruplex-binding properties. However, despite their in vitro efficacy, these peptides showed limited cell penetration and anticancer activity. To overcome this challenge, cell-penetrating peptide (CPP)-conjugated derivatives were designed, some of which exhibited significant cytotoxic effects on cancer cells. Interestingly, selected CPP-conjugated peptides exerted potent anticancer activity across various tumour types via a G-quadruplex-dependent mechanism. These findings underscore the potential of peptide-based G-quadruplex ligands in cancer therapy and pave the way for the development of novel therapeutic strategies targeting these DNA structures.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , G-Quadruplexes , G-Quadruplexes/drug effects , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Cell Line, Tumor , Peptides/chemistry , Peptides/pharmacology , Ligands , DNA/chemistry , DNA/metabolism , Shelterin Complex/metabolism , Shelterin Complex/chemistry , Protein Binding
13.
Eur J Med Chem ; 274: 116536, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38805936

ABSTRACT

G-quadruplexes (G4s) are commonly formed in the G-rich strand of telomeric DNA. Ligands targeting telomeric G4 induce DNA damage and telomere dysfunction, which makes them potential antitumor drugs. New telomeric G4 ligands with drug-likeness are still needed to be exploited, especially with their antitumor mechanisms thoroughly discussed. In this study, a novel series of quinoxaline analogs were rationally designed and synthesized. Among them, R1 was the most promising ligand for its cytotoxic effects on tumor cells and stabilizing ability with telomeric G4. Cellular assays illustrated that R1 stabilized G4 and induced R-loop accumulation in the telomeric regions, subsequently triggering DNA damage responses, cell cycle arrest in G2/M phase, apoptosis and antiproliferation. Moreover, R1 evoked immunogenic cell death (ICD) in tumor cells, which promoted the maturation of bone marrow derived dendritic cells (BMDCs). In breast cancer mouse model, R1 exhibited a significant decrease in tumor burden through the immunomodulatory effects, including the increase of CD4+ and CD8+ T cells in tumors and cytokine levels in sera. Our research provides a new idea that targeting telomeric G4 induces DNA damage responses, causing antitumor effects both in vitro and in vivo, partially due to the enhancement of immunomodulation.


Subject(s)
Antineoplastic Agents , Cell Proliferation , G-Quadruplexes , Quinoxalines , Telomere , G-Quadruplexes/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Quinoxalines/chemistry , Quinoxalines/pharmacology , Quinoxalines/chemical synthesis , Animals , Humans , Telomere/drug effects , Ligands , Mice , Cell Proliferation/drug effects , Molecular Structure , Drug Screening Assays, Antitumor , Structure-Activity Relationship , Dose-Response Relationship, Drug , Female , Immunomodulation/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Mice, Inbred BALB C , DNA Damage/drug effects
14.
Bioorg Chem ; 148: 107475, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38772293

ABSTRACT

The applications of antisense oligonucleotides (ASOs) in rare or common diseases treatment have garnered great attention in recent years. Nevertheless, challenges associated with stability and bioavailability still persist, hampering the efficiency of ASOs. This work presents an ASO prodrug with parallel G-quadruplex assembly and lysosome escape capabilities for oncotherapy. Our findings revealed that the end-assembled quadruplex structure effectively shielded the ASO from enzymatic degradation. Meanwhile, the conjugation of maleimide within the quadruplex enhanced cellular uptake, potentially offering an alternative cell entry mechanism that circumvents lysosome involvement. Notably, an optimized molecule, Mal2-G4-ASO, exhibited remarkable therapeutic effects both in vitro and in vivo. This work presents a promising avenue for enhancing the activity of nucleic acid drugs in oncotherapy and potentially other disease contexts.


Subject(s)
G-Quadruplexes , Lysosomes , Oligonucleotides, Antisense , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , G-Quadruplexes/drug effects , Humans , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/chemical synthesis , Lysosomes/metabolism , Animals , Molecular Structure , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mice , Drug Screening Assays, Antitumor , Dose-Response Relationship, Drug , Cell Proliferation/drug effects , Structure-Activity Relationship , Cell Line, Tumor , Mice, Nude , Mice, Inbred BALB C
15.
J Biomed Sci ; 31(1): 50, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741159

ABSTRACT

BACKGROUND: G-quadruplex DNA (G4) is a non-canonical structure forming in guanine-rich regions, which play a vital role in cancer biology and are now being acknowledged in both nuclear and mitochondrial (mt) genome. However, the impact of G4-based targeted therapy on both nuclear and mt genome, affecting mt function and its underlying mechanisms remain largely unexplored. METHODS: The mechanisms of action and therapeutic effects of a G4-binding platinum(II) complex, Pt-ttpy, on mitochondria were conducted through a comprehensive approaches with in vitro and in vivo models, including ICP-MS for platinum measurement, PCR-based genetic analysis, western blotting (WB), confocal microscope for mt morphology study, extracellular flux analyzer, JC1 and Annexin V apoptosis assay, flow cytometry and high content microscope screening with single-cell quantification of both ROS and mt specific ROS, as well as click-chemistry for IF study of mt translation. Decipher Pt-ttpy effects on nuclear-encoded mt related genes expression were undertaken via RNA-seq, Chip-seq and CUT-RUN assays. RESULTS: Pt-ttpy, shows a highest accumulation in the mitochondria of A2780 cancer cells as compared with two other platinum(II) complexes with no/weak G4-binding properties, Pt-tpy and cisplatin. Pt-ttpy induces mtDNA deletion, copy reduction and transcription inhibition, hindering mt protein translation. Functional analysis reveals potent mt dysfunction without reactive oxygen species (ROS) induction. Mechanistic study provided first evidence that most of mt ribosome genes are highly enriched in G4 structures in their promoter regions, notably, Pt-ttpy impairs most nuclear-encoded mt ribosome genes' transcription through dampening the recruiting of transcription initiation and elongation factors of NELFB and TAF1 to their promoter with G4-enriched sequences. In vivo studies show Pt-ttpy's efficient anti-tumor effects, disrupting mt genome function with fewer side effects than cisplatin. CONCLUSION: This study underscores Pt-ttpy as a G4-binding platinum(II) complex, effectively targeting cancer mitochondria through dual action on mt and nuclear G4-enriched genomes without inducing ROS, offering promise for safer and effective platinum-based G4-targeted cancer therapy.


Subject(s)
G-Quadruplexes , Mitochondria , G-Quadruplexes/drug effects , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Genome, Mitochondrial , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Platinum/pharmacology , Animals
16.
Adv Healthc Mater ; 13(20): e2400256, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38669674

ABSTRACT

Cancer is indisputably one of the major threats to mankind, and hence the design of new approaches for the improvement of existing therapeutic strategies is always wanted. Herein, the design of a tumor microenvironment-responsive, DNA-based chemodynamic therapy (CDT) nanoagent with dual Fenton reaction centers for targeted cancer therapy is reported. Self-assembly of DNA amphiphile containing copper complex as the hydrophobic Fenton reaction center results in the formation of CDT-active DNAsome with Cu2+-based Fenton catalytic site as the hydrophobic core and hydrophilic ssDNA protrude on the surface. DNA-based surface addressability of the DNAsome is then used for the integration of second Fenton reaction center, which is a peroxidase-mimicking DNAzyme noncovalently loaded with Hemin and Doxorubicin, via DNA hybridization to give a CDT agent having dual Fenton reaction centers. Targeted internalization of the CDT nanoagent and selective generation of •OH inside HeLa cell are also shown. Excellent therapeutic efficiency is observed for the CDT nanoagent both in vitro and in vivo, and the enhanced efficacy is attributed to the combined and synergetic action of CDT and chemotherapy.


Subject(s)
DNA, Catalytic , Doxorubicin , Humans , HeLa Cells , Doxorubicin/chemistry , Doxorubicin/pharmacology , DNA, Catalytic/chemistry , DNA, Catalytic/metabolism , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , G-Quadruplexes/drug effects , Mice , Copper/chemistry , Tumor Microenvironment/drug effects , Mice, Nude
17.
J Med Chem ; 67(9): 7006-7032, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38668707

ABSTRACT

G-quadruplexes are noncanonical four-stranded DNA secondary structures. MYC is a master oncogene and the G-quadruplex formed in the MYC promoter functions as a transcriptional silencer and can be stabilized by small molecules. We have previously revealed a novel mechanism of action for indenoisoquinoline anticancer drugs, dual-downregulation of MYC and inhibition of topoisomerase I. Herein, we report the design and synthesis of novel 7-aza-8,9-methylenedioxyindenoisoquinolines based on desirable substituents and π-π stacking interactions. These compounds stabilize the MYC promoter G-quadruplex, significantly lower MYC levels in cancer cells, and inhibit topoisomerase I. MYC targeting was demonstrated by differential activities in Raji vs CA-46 cells and cytotoxicity in MYC-dependent cell lines. Cytotoxicities in the NCI-60 panel of human cancer cell lines were investigated. Favorable pharmacokinetics were established, and in vivo anticancer activities were demonstrated in xenograft mouse models. Furthermore, favorable brain penetration, brain pharmacokinetics, and anticancer activity in an orthotopic glioblastoma mouse model were demonstrated.


Subject(s)
Antineoplastic Agents , Drug Design , G-Quadruplexes , Isoquinolines , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc , Topoisomerase I Inhibitors , G-Quadruplexes/drug effects , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Isoquinolines/pharmacology , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Isoquinolines/chemical synthesis , Mice , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/pharmacokinetics , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/therapeutic use , Structure-Activity Relationship , DNA Topoisomerases, Type I/metabolism , Xenograft Model Antitumor Assays
18.
Eur J Med Chem ; 271: 116406, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38688064

ABSTRACT

NRAS mutation is the second most common oncogenic factor in cutaneous melanoma. Inhibiting NRAS translation by stabilizing the G-quadruplex (G4) structure with small molecules seems to be a potential strategy for cancer therapy due to the NRAS protein's lack of a druggable pocket. To enhance the effects of previously reported G4 stabilizers quindoline derivatives, we designed and synthesized a novel series of quindoline derivatives with fork-shaped side chains by introducing (alkylamino)alkoxy side chains. Panels of experimental results showed that introducing a fork-shaped (alkylamino)alkoxy side chain could enhance the stabilizing abilities of the ligands against NRAS RNA G-quadruplexes and their anti-melanoma activities. One of them, 10b, exhibited good antitumor activity in the NRAS-mutant melanoma xenograft mouse model, showing the therapeutic potential of this kind of compounds.


Subject(s)
Antineoplastic Agents , Drug Design , G-Quadruplexes , GTP Phosphohydrolases , Membrane Proteins , G-Quadruplexes/drug effects , Humans , Animals , GTP Phosphohydrolases/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Membrane Proteins/genetics , Structure-Activity Relationship , Molecular Structure , Melanoma/drug therapy , Melanoma/pathology , Dose-Response Relationship, Drug , Cell Proliferation/drug effects , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis , Cell Line, Tumor , Drug Screening Assays, Antitumor , RNA/metabolism , RNA/chemistry , Protein Biosynthesis/drug effects , Alkaloids , Quinolines
19.
J Med Chem ; 67(8): 6292-6312, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38624086

ABSTRACT

Mitochondria are important drug targets for anticancer and other disease therapies. Certain human mitochondrial DNA sequences capable of forming G-quadruplex structures (G4s) are emerging drug targets of small molecules. Despite some mitochondria-selective ligands being reported for drug delivery against cancers, the ligand design is mostly limited to the triphenylphosphonium scaffold. The ligand designed with lipophilic small-sized scaffolds bearing multipositive charges targeting the unique feature of high mitochondrial membrane potential (MMP) is lacking and most mitochondria-selective ligands are not G4-targeting. Herein, we report a new small-sized dicationic lipophilic ligand to target MMP and mitochondrial DNA G4s to enhance drug delivery for anticancer. The ligand showed marked alteration of mitochondrial gene expression and substantial induction of ROS production, mitochondrial dysfunction, DNA damage, cellular senescence, and apoptosis. The ligand also exhibited high anticancer activity against HCT116 cancer cells (IC50, 3.4 µM) and high antitumor efficacy in the HCT116 tumor xenograft mouse model (∼70% tumor weight reduction).


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , G-Quadruplexes , Mitochondria , Humans , G-Quadruplexes/drug effects , Ligands , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Mice , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Apoptosis/drug effects , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Mice, Nude , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Xenograft Model Antitumor Assays , HCT116 Cells , DNA, Mitochondrial/metabolism
20.
Int J Biol Macromol ; 269(Pt 1): 131806, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670179

ABSTRACT

Acinetobacter baumannii is a notorious pathogen that commonly thrives in hospital environments and is responsible for numerous nosocomial infections in humans. The burgeoning multi-drug resistance leaves relatively minimal options for treating the bacterial infection, posing a significant problem and prompting the identification of new approaches for tackling the same. This motivated us to focus on non-canonical nucleic acid structures, mainly G-quadruplexes, as drug targets. G-quadruplexes have recently been gaining attention due to their involvement in multiple bacterial and viral pathogenesis. Herein, we sought to explore conserved putative G-quadruplex motifs in A. baumannii. In silico analysis revealed the presence of eight conserved motifs in genes involved in bacterial survival and pathogenesis. The biophysical and biomolecular analysis confirmed stable G-quadruplex formation by the motifs and showed a high binding affinity with the well-reported G-quadruplex binding ligand, BRACO-19. BRACO-19 exposure also decreased the growth of bacteria and downregulated the expression of G-quadruplex-harboring genes. The biofilm-forming ability of the bacteria was also affected by BRACO-19 addition. Taking all these observations into account, we have shown here for the first time the potential of G-quadruplex structures as a promising drug target in Acinetobacter baumannii, for addressing the challenges posed by this infamous pathogen.


Subject(s)
Acinetobacter baumannii , G-Quadruplexes , Acinetobacter baumannii/genetics , Acinetobacter baumannii/drug effects , G-Quadruplexes/drug effects , Biofilms/drug effects , Biofilms/growth & development , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Gene Expression Regulation, Bacterial/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL