Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 383
Filter
1.
Front Cell Infect Microbiol ; 14: 1414135, 2024.
Article in English | MEDLINE | ID: mdl-38863831

ABSTRACT

Introduction: Acanthamoeba infection is a serious public health concern, necessitating the development of effective and safe anti-Acanthamoeba chemotherapies. Poly (ADP-ribose) polymerases (PARPs) govern a colossal amount of biological processes, such as DNA damage repair, protein degradation and apoptosis. Multiple PARP-targeted compounds have been approved for cancer treatment. However, repurposing of PARP inhibitors to treat Acanthamoeba is poorly understood. Methods: In the present study, we attempted to fill these knowledge gaps by performing anti-Acanthamoeba efficacy assays, cell biology experiments, bioinformatics, and transcriptomic analyses. Results: Using a homology model of Acanthamoeba poly (ADP-ribose) polymerases (PARPs), molecular docking of approved drugs revealed three potential inhibitory compounds: olaparib, venadaparib and AZ9482. In particular, venadaparib exhibited superior docking scores (-13.71) and favorable predicted binding free energy (-89.28 kcal/mol), followed by AZ9482, which showed a docking score of -13.20 and a binding free energy of -92.13 kcal/mol. Notably, the positively charged cyclopropylamine in venadaparib established a salt bridge (through E535) and a hydrogen bond (via N531) within the binding pocket. For comparison, AZ9482 was well stacked by the surrounding aromatic residues including H625, Y652, Y659 and Y670. In an assessment of trophozoites viability, AZ9482 exhibited a dose-and time-dependent anti-trophozoite effect by suppressing Acanthamoeba PARP activity, unlike olaparib and venadaparib. An Annexin V-fluorescein isothiocyanate/propidium iodide apoptosis assay revealed AZ9482 induced trophozoite necrotic cell death rather than apoptosis. Transcriptomics analyses conducted on Acanthamoeba trophozoites treated with AZ9482 demonstrated an atlas of differentially regulated proteins and genes, and found that AZ9482 rapidly upregulates a multitude of DNA damage repair pathways in trophozoites, and intriguingly downregulates several virulent genes. Analyzing gene expression related to DNA damage repair pathway and the rate of apurinic/apyrimidinic (AP) sites indicated DNA damage efficacy and repair modulation in Acanthamoeba trophozoites following AZ9482 treatment. Discussion: Collectively, these findings highlight AZ9482, as a structurally unique PARP inhibitor, provides a promising prototype for advancing anti-Acanthamoeba drug research.


Subject(s)
Molecular Docking Simulation , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Humans , Piperazines/pharmacology , Phthalazines/pharmacology , Phthalazines/chemistry , Drug Repositioning , Poly(ADP-ribose) Polymerases/metabolism , Acanthamoeba/drug effects , Computational Biology , Apoptosis/drug effects , Gene Expression Profiling , Antiprotozoal Agents/pharmacology , Trophozoites/drug effects
2.
Chem Res Toxicol ; 37(6): 1023-1034, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38743824

ABSTRACT

The clinically used antihypertensive agent hydralazine rapidly generates hydrazone-derived adducts by reaction with apurinic/apyrimidinic (also known as abasic or AP) sites in many different sequences of duplex DNA. The reaction rates are comparable to those of some AP-trapping reagents previously described as "ultrafast." Initially, reversible formation of a hydrazone adduct is followed by an oxidative cyclization reaction that generates a chemically stable triazolo[3,4-a]phthalazine adduct. The net result is that the reaction of hydralazine with AP sites in duplex DNA yields a rapid and irreversible adduct formation. Although the hydrazone and triazolo[3,4-a]phthalazine adducts differ by only two mass units, it was possible to use MALDI-TOF-MS and ESI-QTOF-nanospray-MS to quantitatively characterize mixtures of these adducts by deconvolution of overlapping isotope envelopes. Reactions of hydralazine with the endogenous ketone pyruvate do not prevent the formation of the hydralazine-AP adducts, providing further evidence that these adducts have the potential to form in cellular DNA. AP sites are ubiquitous in cellular DNA, and rapid, irreversible adduct formation by hydralazine could be relevant to the pathogenesis of systemic drug-induced lupus erythematosus experienced by some patients. Finally, hydralazine might be developed as a probe for the detection of AP sites, the study of cellular BER, and marking the location of AP sites in DNA-sequencing analyses.


Subject(s)
DNA Adducts , DNA , Hydralazine , Phthalazines , Hydralazine/chemistry , DNA/chemistry , DNA/drug effects , DNA Adducts/chemistry , Phthalazines/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Antihypertensive Agents/chemistry , Triazoles/chemistry , Spectrometry, Mass, Electrospray Ionization
3.
J Med Chem ; 67(11): 8877-8901, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38776379

ABSTRACT

Designing selective PARP-1 inhibitors has become a new strategy for anticancer drug development. By sequence comparison of PARP-1 and PARP-2, we identified a possible selective site (S site) consisting of several different amino acid residues of α-5 helix and D-loop. Targeting this S site, 140 compounds were designed, synthesized, and characterized for their anticancer activities and mechanisms. Compound I16 showed the highest PARP-1 enzyme inhibitory activity (IC50 = 12.38 ± 1.33 nM) and optimal selectivity index over PARP-2 (SI = 155.74). Oral administration of I16 (25 mg/kg) showed high inhibition rates of Hela and SK-OV-3 tumor cell xenograft models, both of which were higher than those of the oral positive drug Olaparib (50 mg/kg). In addition, I16 has an excellent safety profile, without significant toxicity at high oral doses. These findings provide a novel design strategy and chemotype for the development of safe, efficient, and highly selective PARP-1 inhibitors.


Subject(s)
Antineoplastic Agents , Drug Design , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Mice , Structure-Activity Relationship , Cell Line, Tumor , Mice, Nude , Female , Xenograft Model Antitumor Assays , HeLa Cells , Molecular Docking Simulation , Mice, Inbred BALB C , Cell Proliferation/drug effects , Phthalazines/pharmacology , Phthalazines/chemistry , Phthalazines/chemical synthesis
4.
Eur J Med Chem ; 240: 114574, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35785724

ABSTRACT

Based on the reported synthetic lethality of the combination of PARP inhibitor olaparib with the natural product alantolactone, we designed several series of new PARP1 inhibitors by structurally merging both compounds into a single hybrid compound. Among them, compounds 20e and 25a displayed not only high biochemical activity (IC50 = 2.99 nM and 5.91 nM vs 11.36 nM), but also higher inhibitory effects against proliferation of BRCA1-deficient UWB1.289 cells than olaparib (IC50 = 0.27 µM and 0.41 µM vs 0.66 µM). Much weak activity was observed in BRCA1 wild-type human fetal lung IMR-90 and WI-38 cells (IC50s > 10 µM). Treatment with compounds 20e and 25a was found to induce increased levels of γH2AX in a concentration-dependent manner in both MDA-MB-436 and Capan-1 cells to a degree comparable with that of olaparib. Further mechanism study indicated that these compounds activated the cell cycle checkpoints, and subsequently induced G2/M arrest and apoptosis. The results validated that merging PARP inhibitors with other DNA-damage related compounds would produce more potent PARP inhibitors for anticancer studies. However, the poor aqueous solubility and low cell penetration of the current hybrid compounds call for further structural optimization.


Subject(s)
Biological Products , Poly(ADP-ribose) Polymerase Inhibitors , Apoptosis , Biological Products/pharmacology , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Humans , Lactones , Phthalazines/chemistry , Piperazines , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Sesquiterpenes, Eudesmane
5.
Biotechnol Appl Biochem ; 69(5): 2017-2027, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34622994

ABSTRACT

A new method for the synthesis of 1H-pyrazolo[1,2-b]phthalazine-5,10-dione derivatives via lipase from the Aspergillus niger-catalyzed multicomponent reaction of aldehydes, malononitrile, and phthalhydrazide is reported herein for the first time. This novel method holds several advantages, including its efficiency, environmental friendliness, simple workup procedure, and good yield (70-86%). The effects of temperature, organic solvents, and water content were investigated. This protocol has the potential to replace traditional chemical synthesis routes for the synthesis of nitrogen-containing heterocyclic compounds.


Subject(s)
Lipase , Phthalazines , Solvents/chemistry , Phthalazines/chemistry , Water/chemistry
6.
Arch Pharm (Weinheim) ; 355(1): e2100278, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34596910

ABSTRACT

Twenty new N-substituted-4-phenylphthalazin-1-amine derivatives were designed, synthesized, and evaluated for their anticancer activities against HepG2, HCT-116, and MCF-7 cells as VEGFR-2 inhibitors. HCT-116 was the most sensitive cell line to the influence of the new derivatives. In particular, compound 7f was found to be the most potent derivative among all the tested compounds against the three cancer cell lines, with 50% inhibition concentration, IC50 = 3.97, 4.83, and 4.58 µM, respectively, which is more potent than both sorafenib (IC50 = 9.18, 5.47, and 7.26 µM, respectively) and doxorubicin (IC50 = 7.94, 8.07, and 6.75 µM, respectively). Fifteen of the synthesized derivatives were selected to evaluate their inhibitory activities against VEGFR-2. Compound 7f was found to be the most potent derivative that inhibited VEGFR-2 at an IC50 value of 0.08 µM, which is more potent than sorafenib (IC50 = 0.10 µM). Compound 8c inhibited VEGFR-2 at an IC50 value of 0.10 µM, which is equipotent to sorafenib. Moreover, compound 7a showed very good activity with IC50 values of 0.11 µM, which is nearly equipotent to sorafenib. In addition, compounds 7d, 7c, and 7g possessed very good VEGFR-2-inhibitory activity, with IC50 values of 0.14, 0.17, and 0.23 µM, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Phthalazines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , HCT116 Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Docking Simulation , Phthalazines/chemical synthesis , Phthalazines/chemistry , Sorafenib/pharmacology , Structure-Activity Relationship
7.
Cell Chem Biol ; 29(2): 202-214.e7, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34329582

ABSTRACT

PARP inhibitors (PARPis) display single-agent anticancer activity in small cell lung cancer (SCLC) and other neuroendocrine tumors independent of BRCA1/2 mutations. Here, we determine the differential efficacy of multiple clinical PARPis in SCLC cells. Compared with the other PARPis rucaparib, olaparib, and niraparib, talazoparib displays the highest potency across SCLC, including SLFN11-negative cells. Chemical proteomics identifies PARP16 as a unique talazoparib target in addition to PARP1. Silencing PARP16 significantly reduces cell survival, particularly in combination with PARP1 inhibition. Drug combination screening reveals talazoparib synergy with the WEE1/PLK1 inhibitor adavosertib. Global phosphoproteomics identifies disparate effects on cell-cycle and DNA damage signaling thereby illustrating underlying mechanisms of synergy, which is more pronounced for talazoparib than olaparib. Notably, silencing PARP16 further reduces cell survival in combination with olaparib and adavosertib. Together, these data suggest that PARP16 contributes to talazoparib's overall mechanism of action and constitutes an actionable target in SCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Aged , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Drug Screening Assays, Antitumor , Female , Humans , Male , Phthalazines/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
8.
J Enzyme Inhib Med Chem ; 37(1): 299-314, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34894955

ABSTRACT

This research presents the design and synthesis of a novel series of phthalazine derivatives as Topo II inhibitors, DNA intercalators, and cytotoxic agents. In vitro testing of the new compounds against HepG-2, MCF-7, and HCT-116 cell lines confirmed their potent cytotoxic activity with low IC50 values. Topo II inhibition and DNA intercalating activities were evaluated for the most cytotoxic members. IC50 values determination demonstrated Topo II inhibitory activities and DNA intercalating affinities of the tested compounds at a micromolar level. Amongst, compound 9d was the most potent member. It inhibited Topo II enzyme at IC50 value of 7.02 ± 0.54 µM with DNA intercalating IC50 of 26.19 ± 1.14 µM. Compound 9d was then subjected to an in vivo antitumor examination. It inhibited tumour proliferation reducing solid tumour volume and mass. Additionally, it restored liver enzymes, proteins, and CBC parameters near-normal, indicating a remarkable amelioration in their functions along with histopathological examinations.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA/chemistry , Drug Design , Molecular Docking Simulation , Phthalazines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phthalazines/chemical synthesis , Phthalazines/chemistry , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Tumor Cells, Cultured
9.
Eur J Med Chem ; 229: 114058, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34954595

ABSTRACT

In view of histone deacetylases (HDACs) as a promising target for cancer therapy, a series of phthalazino[1,2-b]-quinazolinone units were hybrided with ortho-aminoanilide or hydroxamic acid to serve as multi-target HDAC inhibitors for the treatment of solid tumors. Among the target compounds, 8h possessed nano-molar IC50 values toward the tested cancer cells and HDAC subtypes, which was more potent than the HDAC inhibitor SAHA (vorinostat). Mechanism study revealed that compound 8h could suppress the HepG2 cell proliferation via prompting the acetylation of histone 3 (H3) and α-tubulin, and activating the p53 signal pathway as designed. In addition, compound 8h exhibited much stronger in vivo antitumor efficacy than SAHA in the HepG2 xenograft tumor model with negligible toxicity. As a novel multi-target HDAC inhibitor, compound 8h deserves further development as a potential anticancer agent.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Histone Deacetylase Inhibitors , Histone Deacetylases , Liver Neoplasms , Phthalazines , Quinazolinones , Tumor Suppressor Protein p53 , Animals , Humans , Male , Mice , Acetylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Hydroxamic Acids/chemistry , Liver Neoplasms/drug therapy , Molecular Docking Simulation , Phthalazines/chemistry , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , Signal Transduction , Structure-Activity Relationship , Tubulin/metabolism , Tumor Suppressor Protein p53/metabolism , Vorinostat/chemistry
10.
J Med Chem ; 64(21): 15690-15701, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34672571

ABSTRACT

Given the clinical potential of poly(ADP-ribose) polymerases (PARP) imaging for the detection and stratification of various cancers, the development of novel PARP imaging probes with improved pharmacological profiles over established PARP imaging agents is warranted. Here, we present a novel 18F-labeled PARP radiotracer based on the clinically superior PARP inhibitor talazoparib. An automated radiosynthesis of [18F]talazoparib (RCY: 13 ± 3.4%; n = 4) was achieved using a "design of experiments" (DoE) optimized copper-mediated radiofluorination reaction. The chiral product was isolated from the reaction mixture using 2D reversed-phase/chiral radio-HPLC (>99% ee). (8S,9R)-[18F]Talazoparib demonstrated PARP binding in HCC1937 cells in vitro and showed an excellent tumor-to-blood ratio in xenograft-bearing mice (10.2 ± 1.5). Additionally, a favorable pharmacological profile in terms of excretion, metabolism, and target engagement was observed. This synthesis of [18F]talazoparib exemplifies how DoE can enable the radiosyntheses of synthetically challenging radiolabeled compounds of high interest to the imaging community.


Subject(s)
Antineoplastic Agents/pharmacology , Automation , Breast Neoplasms/drug therapy , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Fluorine Radioisotopes , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred NOD , Molecular Structure , Phthalazines/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerases/analysis , Tumor Cells, Cultured
11.
Adv Mater ; 33(45): e2104594, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34554623

ABSTRACT

Inspired by natural saccharide-protein complexes, a stimuli-responsive biodegradable and branched glycopolymer-pyropheophorbide-a (Ppa) conjugate (BSP) with saccharide units for cancer therapy is constructed. A linear glycopolymeric conjugate (LSP), a branched glycopolymeric conjugate (BShP) from Ppa with long carbon chains, and a branched conjugate (BHSP) based on poly[N-(2-hydroxypropyl) methacrylamide] (polyHPMA) without saccharide units are prepared as controls. Through structure-activity relationship studies, BSP with a 3D network structure forms stable nanostructures via weak intermolecular interactions, regulating the stacking state of Ppa to improve the singlet oxygen quantum yield and the corresponding photodynamic therapy (PDT) effect. BSP shows high loading of olaparib, and are further coated with tumor cell membranes, resulting in a biomimetic nanomedicine (CM-BSPO). CM-BSPO shows highly efficient tumor targeting and cellular internalization properties. The engulfment of CM-BSPO accompanied with laser irradiation results in a prominent antitumor effect, evidenced by disruption of cell cycles in tumor cells, increased apoptosis and DNA damage, and subsequent inhibition of repair for damaged DNA. The mechanism for the synergistic effect from PDT and olaparib is unveiled at the genetic and protein level through transcriptome analysis. Overall, this biodegradable and branched glycopolymer-drug conjugate could be effectively optimized as a biomimetic nanomedicine for cancer therapy.


Subject(s)
Biomimetic Materials/chemistry , Genomic Instability , Nanomedicine , Polysaccharides/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , DNA Damage/drug effects , Drug Carriers/chemistry , Genomic Instability/drug effects , Humans , Light , Mice , Nanostructures/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Phthalazines/chemistry , Phthalazines/metabolism , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Polymethacrylic Acids/chemistry , Reactive Oxygen Species/metabolism
12.
J Med Chem ; 64(17): 12469-12486, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34459195

ABSTRACT

Designing hybrid molecules with dual functions is one approach to improve the therapeutic efficacy of combination treatment. We have previously conjugated phthalazine and bis(hydroxymethyl)pyrrole pharmacophores to form hybrids bearing antiangiogenesis and DNA interstrand cross-linking activities. To improve the bioavailability, we adopted a benzology approach to design and synthesize a new series of 1,2-bis(hydroxymethyl)benzo[g]pyrrolo[2,1-a]phthalazines. These new hybrids retained the dual functions and could be formulated into vehicles for intravenous and oral administration. Among them, we demonstrated that compound 19a with dimethylamine at the C6 position markedly suppressed the tumor growth of human small cell lung cancer cell line H526, squamous lung cancer cell line H520, and renal cancer cell line 786-O in nude mice, implying that compound 19a is a broad-spectrum anticancer agent. Our results implicated that the conjugation of antiangiogenic and DNA cross-linking is likely to be a helpful approach to improving the efficacy of combination therapy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/prevention & control , Phthalazines/chemistry , Phthalazines/pharmacology , Animals , Cell Line, Tumor , Cell Survival , Drug Design , Humans , Lung Neoplasms , Mice , Mice, Nude , Neoplasms, Squamous Cell , Small Cell Lung Carcinoma , Structure-Activity Relationship , Xenograft Model Antitumor Assays
13.
Arch Pharm (Weinheim) ; 354(11): e2100201, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34411344

ABSTRACT

In the designed compounds, a new linker was inserted in the form of fragments with verified VEGFR-2 inhibitory potential, including an α,ß-unsaturated ketonic fragment, pyrazole, and pyrimidine. Also, new distal hydrophobic moieties were attached to these linkers that are expected to increase the hydrophobic interaction with VEGFR-2 and, consequently, the affinity. These structural optimizations have led us to identify the novel dihydropyrazole derivative 6e as a promising hit molecule. All the new derivatives were evaluated to assess their anticancer activity against three human cancer cell lines, including HepG2, HCT-116, and MCF-7. The results of the in vitro anticancer evaluation study revealed the moderate to excellent cytotoxicity of 6c , 6e , 6g , and 7b , with IC50 values in the low micromolar range. The inhibitory activity of VEGFR-2 was investigated for 16 of the designed compounds. The enzyme assay results of the new compounds were compared with those of sorafenib as a reference VEGFR-2 inhibitor. The obtained results demonstrated that our derivatives are potent VEGFR-2 inhibitors. The most potent derivatives 6c , 6e , 6g , and 7b showed IC50 values in the range of 0.11-0.22 µM. Molecular docking and pharmacokinetic studies were also conducted to rationalize the VEGFR-2 inhibitory activity and to evaluate the ability of the most potent derivatives to be developed as good drug candidates.


Subject(s)
Antineoplastic Agents/pharmacology , Phthalazines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , HCT116 Cells , Hep G2 Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Molecular Docking Simulation , Phthalazines/chemical synthesis , Phthalazines/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
14.
Biomolecules ; 11(7)2021 07 03.
Article in English | MEDLINE | ID: mdl-34356606

ABSTRACT

The genetic principle of synthetic lethality has most successfully been exploited in therapies engaging Poly-ADP-ribose-polymerase (PARP) inhibitors to treat patients with homologous recombination (HR)-defective tumors. In this work, we went a step further following the idea of a local molecular cooperation and designed hybrid compounds M1-M3. The drug conjugates M1-M3 combine Olaparib, the first PARP inhibitor approved for clinical use, with Cpd 1, an inhibitor of RAD51 that blocks its HR functions and yet permits RAD51 nucleoprotein filament formation on single-stranded DNA. While in M2 and M3, the parental drugs are linked by -CO-(CH2)n-CO-spacers (n = 2 and 4, respectively), they are directly merged omitting the piperazine ring of Olaparib in M1. Monitoring anti-survival effects of M1-M3 in six breast cancer cell lines of different molecular subtypes showed that in each cell line, at least one of the drug conjugates decreased viability by one to two orders of magnitude compared with parental drugs. While triple-negative breast cancer (TNBC) cells with frequent BRCA1 pathway dysfunction were sensitive to spacer-linked hybrid compounds M1 and M2 regardless of their HR capacities, non-TNBC cells were responsive to the merged drug conjugate M1 only, suggesting different spatial requirements for dual inhibition in these two groups of cell lines. These results demonstrate that, depending on chemical linkage, dual PARP1-RAD51 inhibitory drugs can either sensitize non-TNBC and re-sensitize TNBC cells, or discriminate between these groups of cells.


Subject(s)
Antineoplastic Agents , Neoplasm Proteins/antagonists & inhibitors , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors , Rad51 Recombinase/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Female , Humans , MCF-7 Cells , Neoplasm Proteins/metabolism , Phthalazines/chemistry , Phthalazines/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
15.
Int J Mol Sci ; 22(16)2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34445442

ABSTRACT

DNA repair inhibitors are one of the latest additions to cancer chemotherapy. In general, chemotherapy produces DNA damage but tumoral cells may become resistant if enzymes involved in DNA repair are overexpressed and are able to reverse DNA damage. One of the most successful drugs based on modulating DNA repair are the poly(ADP-ribose) polymerase 1 (PARP1) inhibitors. Several PARP1 inhibitors have been recently developed and approved for clinical treatments. We envisaged that PARP inhibition could be potentiated by simultaneously modulating the expression of PARP 1 and the enzyme activity, by a two-pronged strategy. A noncanonical G-quadruplex-forming sequence within the PARP1 promoter has been recently identified. In this study, we explored the potential binding of clinically approved PARP1 inhibitors to the G-quadruplex structure found at the gene promoter region. The results obtained by NMR, CD, and fluorescence titration confirmed by molecular modeling demonstrated that two out the four PARP1 inhibitors studied are capable of forming defined complexes with the PARP1 G-quadruplex. These results open the possibility of exploring the development of better G-quadruplex binders that, in turn, may also inhibit the enzyme.


Subject(s)
G-Quadruplexes , Models, Molecular , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Promoter Regions, Genetic , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , DNA/chemistry , DNA/drug effects , Humans , Indazoles/chemistry , Indazoles/pharmacology , Magnetic Resonance Spectroscopy , Phthalazines/chemistry , Phthalazines/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
16.
Chem Pharm Bull (Tokyo) ; 69(7): 620-629, 2021.
Article in English | MEDLINE | ID: mdl-34193711

ABSTRACT

Poly(ADP-ribose)polymerase (PARP) is a significant therapeutic target for the treatment of numerous human diseases. Olaparib has been approved as a PARP inhibitor. In this paper, a series of new compounds were designed and synthesized with Olaparib as the lead compound. In order to evaluate the inhibitory activities against PARP1 of the synthesized compounds, in vitro PARP1 inhibition assay and intracellular PARylation assay were conducted. The results showed that the inhibitory activities of the derivatives were related to the type of substituent and the length of alkyl chain connecting the aromatic ring. 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT)-based assay also proved that these compounds demonstrating strong inhibition to PARP1 also have high anti-proliferative activities against BRCA2-deficient cell line (Capan-1). Analysis of the entire results suggest that compound 23 with desirable inhibitory efficiency may hold promise for further in vivo exploration of PARP inhibition.


Subject(s)
Drug Design , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Docking Simulation , Phthalazines/chemical synthesis , Phthalazines/chemistry , Phthalazines/pharmacology , Piperazines/chemical synthesis , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Structure-Activity Relationship
17.
Article in English | MEDLINE | ID: mdl-34233568

ABSTRACT

Although tea is often considered a healthy drink, there is the possibility for it to contain bisphenol A and phthalates. This project was designed to quantitate the amount of these compounds when tea was prepared in a variety of conditions, and with a variety of different brands and flavours. BPA and phthalates were extracted using solid phase extraction and quantitated using gas chromatography - mass spectrometry. The leaching concentration of di-n-butyl phthalate, a major phthalate in dry tea samples, increased with respect to both brewing time and temperature at rates of 5.9 ng/g/min and 2.3 ng/g/°C, respectively. Loose leaf green teas showed lower concentrations of contaminants than bagged teas. The highest concentrations found of all compounds were for benzylbutyl phthalate in both Brand2 English breakfast and Brand2 green teas with concentrations of 244 ± 76 ng/g and 197 ± 9 ng/g, respectively. Di-n-butyl phthalate, benzylbutyl phthalate and bis-2-ethylhexyl phthalate were all present in concentrations of 50 ng/g or more in 3 or more samples.


Subject(s)
Benzhydryl Compounds/chemistry , Phenols/chemistry , Phthalazines/chemistry , Tea/chemistry , Temperature , Environmental Pollutants/chemistry , Estrogens, Non-Steroidal/chemistry , Time Factors
18.
Eur J Med Chem ; 223: 113660, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34246853

ABSTRACT

TGFß is crucial for the homeostasis of epithelial and neural tissues, wound repair, and regulating immune responses. Its dysregulation is associated with a vast number of diseases, of which modifying the tumor microenvironment is one of vital clinical interest. Despite various attempts, there is still no FDA-approved therapy to inhibit the TGFß pathway. Major mainstream approaches involve impairment of the TGFß pathway via inhibition of the TGFßRI kinase. With the purpose to identify non-receptor kinase-based inhibitors to impair TGFß signaling, an in-house chemical library was enriched, through a computational study, to eliminate TGFßRI kinase activity. Selected compounds were screened against a cell line engineered with a firefly luciferase gene under TGFß-Smad-dependent transcriptional control. Results indicated moderate potency for a molecule with phthalazine core against TGFß-Smad signaling. A series of phthalazine compounds were synthesized and evaluated for potency. The most promising compound (10p) exhibited an IC50 of 0.11 ± 0.02 µM and was confirmed to be non-cytotoxic up to 12 µM, with a selectivity index of approximately 112-fold. Simultaneously, 10p was confirmed to reduce the Smad phosphorylation using Western blot without exhibiting inhibition on the TGFßRI enzyme. This study identified a novel small-molecule scaffold that targets the TGFß pathway via a non-receptor-kinase mechanism.


Subject(s)
Phthalazines/chemistry , Transforming Growth Factor beta/antagonists & inhibitors , Cell Survival/drug effects , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Phosphorylation/drug effects , Phthalazines/metabolism , Phthalazines/pharmacology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/chemistry , Smad Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Transforming Growth Factor beta/metabolism
19.
Bioorg Med Chem ; 42: 116266, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34126285

ABSTRACT

Inhibition of PCAF bromodomain has been validated as a promising strategy for the treatment of cancer. In this study, we report the bioisosteric modification of the first reported potent PCAF bromodomain inhibitor, L-45 to its triazoloquinazoline bioisosteres. Accordingly, three new series of triazoloquinazoline derivatives were designed, synthesized, and assessed for their anticancer activity against a panel of four human cancer cells. Three derivatives demonstrated comparable cytotoxic activity with the reference drug doxorubicin. Among them, compound 22 showed the most potent activity with IC50 values of 15.07, 9.86, 5.75, and 10.79 µM against Hep-G2, MCF-7, PC3, and HCT-116 respectively. Also, compound 24 exhibited remarkable cytotoxicity effects against the selected cancer cell lines with IC50 values of 20.49, 12.56, 17.18, and 11.50 µM. Compounds 22 and 25 were the most potent PCAF inhibitors (IC50, 2.88 and 3.19 µM, respectively) compared with bromosporine (IC50, 2.10 µM). Follow up apoptosis induction and cell cycle analysis studies revealed that the bioisostere 22 could induce apoptotic cell death and arrest the cell cycle of PC3 at the G2/M phase. The in silico molecular docking studies were additionally performed to rationalize the PCAF inhibitory effects of new triazoloquinazoline bioisosteres.


Subject(s)
Antineoplastic Agents/pharmacology , Phthalazines/pharmacology , Quinazolines/pharmacology , Triazoles/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Phthalazines/chemical synthesis , Phthalazines/chemistry , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , p300-CBP Transcription Factors/metabolism
20.
J Med Chem ; 64(11): 7839-7852, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34038131

ABSTRACT

Inspired by the success of dual-targeting drugs, especially bispecific antibodies, we propose to combine the concept of proteolysis targeting chimera (PROTAC) and dual targeting to design and synthesize dual PROTAC molecules with the function of degrading two completely different types of targets simultaneously. A library of novel dual-targeting PROTAC molecules has been rationally designed and prepared. A convergent synthetic strategy has been utilized to achieve high synthetic efficiency. These dual PROTAC structures are characterized using trifunctional natural amino acids as star-type core linkers to connect two independent inhibitors and E3 ligands together. In this study, gefitinib, olaparib, and CRBN or VHL E3 ligands were used as substrates to synthesize novel dual PROTACs. They successfully degraded both the epidermal growth factor receptor (EGFR) and poly(ADP-ribose) polymerase (PARP) simultaneously in cancer cells. Being the first successful example of dual PROTACs, this technique will greatly widen the range of application of the PROTAC method and open up a new field for drug discovery.


Subject(s)
Drug Design , ErbB Receptors/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Gefitinib/chemistry , Humans , Ligands , Phthalazines/chemistry , Piperazines/chemistry , Proteasome Endopeptidase Complex/genetics , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...