Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters











Publication year range
1.
Adv Healthc Mater ; 13(20): e2400203, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38774999

ABSTRACT

The limited recapitulation of critical cancer features in 2D cultures causes poor translatability of preclinical results from in vitro assays to in vivo tumor models. This contributes to slow drug development with a low success rate. 3D cultures better recapitulate the tumor microenvironment, enabling more accurate predictions when screening drug candidates and improving the development of chemotherapeutics. Platinum (Pt) (IV) compounds are promising prodrugs designed to reduce the severe systemic toxicity of widely used Food and Drug Administration (FDA)-approved Pt(II) drugs such as cisplatin. Here, this work presents spatiotemporal evaluations in 3D colorectal cancer (CRC) spheroids of mitochondria-targeting Pt(IV) complexes. CRC spheroids provide a greater pathophysiological recapitulation of in vivo tumors than 2D cultures by a marked upregulation of the ABCG2 chemoresistance marker expression. Furthermore, new 3D-staining protocols are introduced to evaluate the real-time decrease in mitochondria membrane potential (ΔΨ) in CRC spheroids, and a Pt-sensing dye to quantify the Pt mitochondrial accumulation. Finally, this work demonstrates a correlation between in vitro results and the efficacy of the compounds in vivo. Overall, the CRC spheroids represent a fast and cost-effective model to assess the behavior of Pt compounds in vitro and predict their translational potential in CRC treatment.


Subject(s)
Colorectal Neoplasms , Spheroids, Cellular , Humans , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor/methods , Cell Line, Tumor , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Membrane Potential, Mitochondrial/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Fluorescence
2.
bioRxiv ; 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38352581

ABSTRACT

Cisplatin is a chemotherapy drug that causes permanent hearing loss by injuring cochlear hair cells. The mechanisms that initiate injury are not fully understood, but mitochondria have emerged as potential mediators of hair cell cytotoxicity. Using in vivo live imaging of hair cells in the zebrafish lateral-line organ expressing a genetically encoded indicator of cumulative mitochondrial activity, we first demonstrate that greater redox history increases susceptibility to cisplatin. Next, we conducted time-lapse imaging to understand dynamic changes in mitochondrial homeostasis and observe elevated mitochondrial and cytosolic calcium that surge prior to hair cell death. Furthermore, using a localized probe that fluoresces in the presence of cisplatin, we show that cisplatin directly accumulates in hair cell mitochondria, and this accumulation occurs before mitochondrial dysregulation and apoptosis. Our findings provide evidence that cisplatin directly targets hair cell mitochondria and support that the mitochondria are integral to cisplatin cytotoxicity in hair cells.

3.
Angew Chem Int Ed Engl ; 63(10): e202317901, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38088924

ABSTRACT

Rising antimicrobial resistance (AMR) and lack of innovation in the antibiotic pipeline necessitate novel approaches to discovering new drugs. Metal complexes have proven to be promising antimicrobial compounds, but the number of studied compounds is still low compared to the millions of organic molecules investigated so far. Lately, machine learning (ML) has emerged as a valuable tool for guiding the design of small organic molecules, potentially even in low-data scenarios. For the first time, we extend the application of ML to the discovery of metal-based medicines. Utilising 288 modularly synthesized ruthenium arene Schiff-base complexes and their antibacterial properties, a series of ML models were trained. The models perform well and are used to predict the activity of 54 new compounds. These displayed a 5.7x higher hit-rate (53.7 %) against methicillin-resistant Staphylococcus aureus (MRSA) compared to the original library (9.4 %), demonstrating that ML can be applied to improve the success-rates in the search of new metalloantibiotics. This work paves the way for more ambitious applications of ML in the field of metal-based drug discovery.


Subject(s)
Coordination Complexes , Methicillin-Resistant Staphylococcus aureus , Ruthenium , Ruthenium/pharmacology , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Coordination Complexes/pharmacology
5.
Angew Chem Int Ed Engl ; 62(50): e202310040, 2023 12 11.
Article in English | MEDLINE | ID: mdl-37621226

ABSTRACT

Antimicrobial resistance, caused by persistent adaptation and growing resistance of pathogenic bacteria to overprescribed antibiotics, poses one of the most serious and urgent threats to global public health. The limited pipeline of experimental antibiotics in development further exacerbates this looming crisis and new drugs with alternative modes of action are needed to tackle evolving pathogenic adaptation. Transition metal complexes can replenish this diminishing stockpile of drug candidates by providing compounds with unique properties that are not easily accessible using pure organic scaffolds. We spotlight four emerging strategies to harness these unique properties to develop new targeted antibacterial agents.


Subject(s)
Coordination Complexes , Transition Elements , Anti-Bacterial Agents/pharmacology , Bacteria , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use
6.
J Am Chem Soc ; 145(11): 6453-6461, 2023 03 22.
Article in English | MEDLINE | ID: mdl-36881731

ABSTRACT

Nitroreductases (NTRs) constitute an important class of oxidoreductase enzymes that have evolved to metabolize nitro-containing compounds. Their unique characteristics have spurred an array of potential uses in medicinal chemistry, chemical biology, and bioengineering toward harnessing nitro caging groups and constructing NTR variants for niche applications. Inspired by how they carry out enzymatic reduction via a cascade of hydride transfer reactions, we sought to develop a synthetic small-molecule NTR system based on transfer hydrogenation mediated by transition metal complexes harnessing native cofactors. We report the first water-stable Ru-arene complex capable of selectively and fully reducing nitroaromatics into anilines in a biocompatible buffered aqueous environment using formate as the hydride source. We further demonstrated its application to activate nitro-caged sulfanilamide prodrug in formate-abundant bacteria, specifically pathogenic methicillin-resistant Staphylococcus aureus. This proof of concept paves the way for a new targeted antibacterial chemotherapeutic approach leveraging on redox-active metal complexes for prodrug activation via bioinspired nitroreduction.


Subject(s)
Coordination Complexes , Methicillin-Resistant Staphylococcus aureus , Prodrugs , Prodrugs/pharmacology , Methicillin-Resistant Staphylococcus aureus/metabolism , Coordination Complexes/pharmacology , Bacteria/metabolism , Nitro Compounds/chemistry , Nitroreductases/metabolism , Formates
7.
Metallomics ; 14(12)2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36271844

ABSTRACT

In research enabling preclinical development and attaining a deeper understanding of the behavior of metallodrugs in cancer cells with acquired resistance, intracellular Pt accumulation could be considered an important biomarker and analytical focus. In this work, Pt accumulation patterns in terms of the number of cells and Pt mass in single cells were precisely defined by using inductively coupled plasma-mass spectrometry (ICP-MS) operating in a fast time-resolved analysis mode. This technique is otherwise known as single-cell (SC)-ICP-MS. By applying the nascent and validated SC-ICP-MS technique, comparisons across three Pt drugs (cisplatin, carboplatin, and oxaliplatin) in the A2780 and A2780cis ovarian cancer cell models could be made. Additional roles of transporters on top of passive diffusion and the drugs' bioactivity could be postulated. The SC-ICP-MS-based observations also served as a cross-validation point to augment preexisting research findings on Pt-resistance mechanisms. Conjectures regarding S and Fe metabolism were also derived based on an additional and direct ICP-MS analysis of endogenous elements. Overall, our work not only confirms the utility of SC-ICP-MS in chemotherapeutic research, but also provided insights into further ICP-MS-based analytical capacities to be developed.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Cisplatin/metabolism , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Cell Line, Tumor , Ovarian Neoplasms/drug therapy , Oxaliplatin , Antineoplastic Agents/chemistry
8.
Inorg Chem ; 61(26): 10167-10181, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35713376

ABSTRACT

Indolo[3,2-d][1]benzazepines (paullones), indolo[3,2-d][2]benzazepines, and indolo[2,3-d][2]benzazepines (latonduines) are isomeric scaffolds of current medicinal interest. Herein, we prepared a small library of novel indolo[3,2-d][2]benzazepine-derived ligands HL1-HL4 and copper(II) complexes 1-4. All compounds were characterized by spectroscopic methods (1H and 13C NMR, UV-vis, IR) and electrospray ionization (ESI) mass spectrometry, while complexes 2 and 3, in addition, by X-ray crystallography. Their purity was confirmed by HPLC coupled with high-resolution ESI mass spectrometry and/or elemental analysis. The stability of compounds in aqueous solutions in the presence of DMSO was confirmed by 1H NMR and UV-vis spectroscopy measurements. The compounds revealed high antiproliferative activity in vitro in the breast cancer cell line MDA-MB-231 and hepatocellular carcinoma cell line LM3 in the low micromolar to nanomolar concentration range. Important structure-activity relationships were deduced from the comparison of anticancer activities of HL1-HL4 and 1-4 with those of structurally similar paullone-derived (HL5-HL7 and 5-7) and latonduine-derived scaffolds (HL8-HL11 and 8-11). The high anticancer activity of the lead drug candidate 4 was linked to reactive oxygen species and endoplasmic reticulum stress induction, which were confirmed by fluorescent microscopy and Western blot analysis.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzazepines/chemistry , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Copper/chemistry , Copper/pharmacology , Crystallography, X-Ray , Ligands , Structure-Activity Relationship
9.
JACS Au ; 1(9): 1348-1354, 2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34604844

ABSTRACT

Pathogenic microorganisms pose a serious threat to global public health due to their persistent adaptation and growing resistance to antibiotics. Alternative therapeutic strategies are required to address this growing threat. Bactericidal antibiotics that are routinely prescribed to treat infections rely on hydroxyl radical formation for their therapeutic efficacies. We developed a redox approach to target bacteria using organotransition metal complexes to mediate the reduction of cellular O2 to H2O2, as a precursor for hydroxyl radicals via Fenton reaction. We prepared a library of 480 unique organoruthenium Schiff-base complexes using a coordination-driven three-component assembly strategy and identified the lead organoruthenium complex Ru1 capable of selectively invoking oxidative stress in Gram-positive bacteria, in particular methicillin-resistant Staphylococcus aureus, via transfer hydrogenation reaction and/or single electron transfer on O2. This strategy paves the way for a targeted antimicrobial approach leveraging on the redox chemistry of organotransition metal complexes to combat drug resistance.

10.
Angew Chem Int Ed Engl ; 60(24): 13405-13413, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33755286

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by an aberrant metabolic phenotype with high metastatic capacity, resulting in poor patient prognoses and low survival rates. We designed a series of novel AuIII cyclometalated prodrugs of energy-disrupting Type II antidiabetic drugs namely, metformin and phenformin. Prodrug activation and release of the metformin ligand was achieved by tuning the cyclometalated AuIII fragment. The lead complex 3met was 6000-fold more cytotoxic compared to uncoordinated metformin and significantly reduced tumor burden in mice with aggressive breast cancers with lymphocytic infiltration into tumor tissues. These effects was ascribed to 3met interfering with energy production in TNBCs and inhibiting associated pro-survival responses to induce deadly metabolic catastrophe.


Subject(s)
Antineoplastic Agents/metabolism , Metformin/metabolism , Prodrugs/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cell Line, Tumor , Coordination Complexes/chemistry , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Gold/chemistry , Humans , Metformin/chemistry , Mice , Molecular Conformation , Phenformin/chemistry , Phenformin/metabolism , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Transplantation, Heterologous , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
11.
Inorg Chem ; 60(3): 1823-1831, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33464875

ABSTRACT

A common challenge in Pt(IV) prodrug design is the limited repertoire of linkers available to connect the Pt(IV) scaffold with the bioactive payload. The commonly employed linkers are either too stable, leading to a linker artifact on the payload upon release, or too unstable, leading to premature release. In this study, we report the synthesis of a new class of Pt(IV) prodrugs using masked self-immolative 4-aminobenzyl linkers for controlled and traceless codrug delivery. Upon reduction of self-immolative Pt(IV) prodrugs, the detached axial ligands undergo decarboxylation and 1,6-elimination for payload release. Introduction of self-immolative linkers conferred good aqueous stability to the Pt(IV) codrug complex. Investigation revealed that efficient 1,6-elimination could be attributed to stabilization of the p-aza-quinone-methide intermediate. In particular, the self-immolative Pt(IV) prodrugs with cinnamate and coumarin derivatives were more potent than the coadministration of cisplatin with an unconjugated cinnamate or coumarin payload in vitro.


Subject(s)
Antineoplastic Agents/chemistry , Cisplatin/chemistry , Organoplatinum Compounds/chemistry , Prodrugs/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Humans , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology
12.
Clin Cancer Res ; 27(7): 1875-1881, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33148667

ABSTRACT

PURPOSE: Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a novel laparoscopic, intraperitoneal chemotherapy delivery technique aiming to improve drug distribution and tissue penetration to treat peritoneal metastases. Thus far, PIPAC oxaliplatin is conducted at an arbitrary dose of 92 mg/m2. We conducted a phase I study to establish safety and tolerability. PATIENTS AND METHODS: We used a 3+3 dose-escalation design of PIPAC oxaliplatin for patients with peritoneal metastases from gastrointestinal tumors, after failure of at least first-line chemotherapy. Dose levels were planned at 45, 60, 90, and 120 mg/m2. RESULTS: This study included 16 patients with 24 PIPAC procedures (8 gastric; 5 colorectal; and 1 gallbladder, pancreas, and appendix cancer each). Median age and peritoneal cancer index (PCI) score were 62 years and 17, respectively. Two patients developed pancreatitis (grade 2 and 3) at 45 mg/m2, necessitating cohort expansion. Another patient developed grade 2 pancreatitis at 90 mg/m2. There were no other dose-limiting toxicities, and the highest-dose cohort (120 mg/m2) tolerated PIPAC well. Pharmacokinetic analyses demonstrated good linearity between dose and maximum concentration (r 2 = 0.95) and AUC (r 2 = 0.99). On the basis of RECIST, 62.5% and 50% had stable disease after one and two PIPAC procedures, respectively. A total of 8 patients underwent two PIPAC procedures, with improvement of median PCI and peritoneal regression grade score from 15 to 12 and 2.5 to 2.0, respectively. CONCLUSIONS: The recommended phase II dose is 120 mg/m2. Future studies should further delineate the efficacy and role of PIPAC oxaliplatin for peritoneal metastases.See related commentary by de Jong et al., p. 1830.


Subject(s)
Gastrointestinal Neoplasms/pathology , Oxaliplatin/administration & dosage , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Aerosols , Aged , Female , Humans , Laparoscopy , Male , Middle Aged , Oxaliplatin/adverse effects , Pancreatitis/chemically induced , Peritoneal Neoplasms/mortality , Prospective Studies
13.
Angew Chem Int Ed Engl ; 60(17): 9264-9269, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33290628

ABSTRACT

Mitochondria have emerged as important targets for cisplatin in cancer therapy. Apart from cisplatin, anticancer Pt complexes based on similar scaffolds have also been developed to target mitochondria. Yet cellular processing of cisplatin or these mitochondria-targeting Pt analogues remained unexplored, largely due to a lack of tools capable of probing these Pt drugs within an intracellular environment. We developed the first mitochondria-targeted fluorescent probe for real-time monitoring of Pt accumulation in mitochondria. We applied the probe to investigate mitochondria as cellular targets for Pt drug complexes and uncovered two distinct pathways whereby these Pt complexes could be delivered to mitochondria after cell entry.


Subject(s)
Antineoplastic Agents/metabolism , Cisplatin/metabolism , Fluorescent Dyes/metabolism , Mitochondria/chemistry , Antineoplastic Agents/chemistry , Cisplatin/chemistry , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Mitochondria/metabolism , Optical Imaging , Spectrometry, Fluorescence
14.
ACS Cent Sci ; 6(10): 1651-1653, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33145405
15.
Biomolecules ; 10(9)2020 09 19.
Article in English | MEDLINE | ID: mdl-32961653

ABSTRACT

Thiosemicarbazones continue to attract the interest of researchers as potential anticancer drugs. For example, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, or triapine, is the most well-known representative of this class of compounds that has entered multiple phase I and II clinical trials. Two new triapine derivatives HL1 and HL2 were prepared by condensation reactions of 2-pyridinamidrazone and S-methylisothiosemicarbazidium chloride with 3-N-(tert-butyloxycarbonyl) amino-pyridine-2-carboxaldehyde, followed by a Boc-deprotection procedure. Subsequent reaction of HL1 and HL2 with CuCl2·2H2O in 1:1 molar ratio in methanol produced the complexes [CuII(HL1)Cl2]·H2O (1·H2O) and [CuII(HL2)Cl2] (2). The reaction of HL2 with Fe(NO3)3∙9H2O in 2:1 molar ratio in the presence of triethylamine afforded the complex [FeIII(L2)2]NO3∙0.75H2O (3∙0.75H2O), in which the isothiosemicarbazone acts as a tridentate monoanionic ligand. The crystal structures of HL1, HL2 and metal complexes 1 and 2 were determined by single crystal X-ray diffraction. The UV-Vis and EPR spectroelectrochemical measurements revealed that complexes 1 and 2 underwent irreversible reduction of Cu(II) with subsequent ligand release, while 3 showed an almost reversible electrochemical reduction in dimethyl sulfoxide (DMSO). Aqueous solution behaviour of HL1 and 1, as well as of HL2 and its complex 2, was monitored as well. Complexes 1-3 were tested against ovarian carcinoma cells, as well as noncancerous embryonic kidney cells, in comparison to respective free ligands, triapine and cisplatin. While the free ligands HL1 and HL2 were devoid of antiproliferative activity, their respective metal complexes showed remarkable antiproliferative activity in a micromolar concentration range. The activity was not related to the inhibition of ribonucleotide reductase (RNR) R2 protein, but rather to cancer cell homeostasis disturbance-leading to the disruption of cancer cell signalling.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Pyridines/chemistry , Thiosemicarbazones/chemistry , Aldehydes/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Crystallography, X-Ray , Electrochemical Techniques/methods , HEK293 Cells , Humans , Molecular Structure , Pyridines/chemical synthesis , Pyridines/pharmacology , Spectrophotometry/methods , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/pharmacology
16.
Angew Chem Int Ed Engl ; 59(43): 19070-19078, 2020 10 19.
Article in English | MEDLINE | ID: mdl-32716112

ABSTRACT

Immunogenic cell death (ICD) is a rare immunostimulatory form of cell death that can improve the clinical outcomes of chemo-immunotherapeutic combination regimens through the establishment of a long-term cancer immunity. None of the clinically used DNA-binding PtII complexes is considered a Type II ICD inducer. We generated a series of PtII -carbene complexes by applying minor structural alterations to the scaffold of a Type II ICD inducer Pt-NHC and compared their efficiency in triggering ICD-related cellular responses and phagocytosis. We successfully identified PlatinER, a novel highly potent PtII candidate with superior ICD properties. Crucially, the magnitude of ICD-associated phagocytosis induced upon exposure of cancer cells to Pt complexes was dependent on the levels of ER-localized reactive oxygen species (ROS) generation, which underpins their mechanisms of action and provides a feasible approach for the design of more effective Type II ICD inducers.


Subject(s)
Antineoplastic Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Cell Line, Tumor , Eukaryotic Initiation Factor-2/metabolism , Humans , Immunogenic Cell Death/drug effects , Organoplatinum Compounds/chemistry , Phagocytosis/drug effects , Proton Magnetic Resonance Spectroscopy , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Electrospray Ionization , eIF-2 Kinase/metabolism
17.
Dalton Trans ; 49(22): 7355-7363, 2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32432621

ABSTRACT

Ovarian cancer is a highly aggressive disease which is treated by surgery and platinum chemotherapy. However, a significant proportion of treated patients develop resistance to platinum treatment resulting in tumor relapse. Acquired platinum resistance has been recently correlated with activation of pro-survival endoplasmic reticulum (ER) stress responses. We hypothesized that Au complexes that induce severe ER stress might counteract pro-survival cellular attempts leading to the ER stress-mediated apoptosis and reduced platinum resistance. In this work, we prepared a series of highly cytotoxic AuI-dialkyldithiocarbamate complexes and investigated their anticancer potential in ovarian cancer cells. Complexes demonstrated surprisingly low stability in chloroform, resulting in the formation of an Au chain polymer, which also displayed excellent cytotoxicity. Lead complex 2 induced oxidative stress and ER stress-mediated p53-independent apoptosis associated with PARP cleavage and cell cycle arrest at G2/M phase. Importantly, 2 caused the surface exposure of calreticulin (CRT), which is the first step in the activation of cellular immunogenic response.


Subject(s)
Antineoplastic Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Organogold Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Phosphines/pharmacology , Thiocarbamates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Molecular Structure , Organogold Compounds/chemical synthesis , Organogold Compounds/chemistry , Ovarian Neoplasms/pathology , Oxidative Stress/drug effects , Phosphines/chemistry , Thiocarbamates/chemistry , Tumor Cells, Cultured
18.
Angew Chem Int Ed Engl ; 59(24): 9314-9318, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32141662

ABSTRACT

The abundance and evolving pathogenic behavior of bacterial microorganisms give rise to antibiotic tolerance and resistance which pose a danger to global public health. New therapeutic strategies are needed to keep pace with this growing threat. We propose a novel approach for targeting bacteria by harnessing formate, a cell metabolite found only in particular bacterial species, to activate an antibacterial prodrug and selectively inhibit their growth. This strategy is premised on transfer hydrogenation reaction on a biorthogonal substrate utilizing native formate as the hydride source as a means of uncaging an antibacterial prodrug. Using coordination-directed 3-component assembly to prepare a library of 768 unique Ru-Arene Schiff-base complexes, we identified several candidates that efficiently reduced sulfonyl azide functional group in the presence of formate. This strategy paves the way for a new approach of targeted antibacterial therapy by exploiting unique bacterial metabolites.


Subject(s)
Anti-Bacterial Agents/metabolism , Formates/metabolism , Prodrugs/metabolism , Ruthenium/chemistry , Anti-Bacterial Agents/pharmacology , Catalysis , Hydrogenation , Schiff Bases/chemistry
19.
Chem Asian J ; 15(9): 1449-1455, 2020 May 04.
Article in English | MEDLINE | ID: mdl-32144847

ABSTRACT

Fluorescence microscopy has emerged as an attractive technique to probe the intracellular processing of Pt-based anticancer compounds. Herein, we reported the first through-bond energy transfer (TBET) fluorescent probe NPR1 designed for sensitive detection and quantitation of PtII complexes. The novel TBET probe was successfully applied for ratiometric fluorescence imaging of anticancer PtII complexes such as cisplatin and JM118 in cells. Capitalizing on the ability of the probe to discriminate between PtII complexes and their PtIV derivatives, the probe was further applied to study the activation of PtIV prodrug complexes that are known to release active PtII species after intracellular reduction.


Subject(s)
Antineoplastic Agents/analysis , Cisplatin/analysis , Fluorescent Dyes/chemistry , Organoplatinum Compounds/analysis , Energy Transfer , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Structure , Optical Imaging
SELECTION OF CITATIONS
SEARCH DETAIL