Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Dalton Trans ; 53(14): 6410-6415, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38501501

ABSTRACT

An asymmetric bi-nuclear copper(II) complex with both cytotoxic and immunogenic activity towards breast cancer stem cells (CSCs) is reported. The bi-nuclear copper(II) complex comprises of two copper(II) centres bound to flufenamic acid and 3,4,7,8-tetramethyl-1,10-phenanthroline. The bi-nuclear copper(II) complex exhibits sub-micromolar potency towards breast CSCs grown in monolayers and three-dimensional cultures. Remarkably, the bi-nuclear copper(II) complex is up to 25-fold more potent toward breast CSC mammospheres than salinomycin (a gold standard anti-breast CSC agent) and cisplatin (a clinically administered metallodrug). Mechanistic studies showed that the bi-nuclear copper(II) complex readily enters breast CSCs, elevates intracellular reactive oxygen species levels, induces apoptosis, and promotes damage-associated molecular pattern release. The latter triggers phagocytosis of breast CSCs by macrophages. As far as we are aware, this is the first report of a bi-nuclear copper(II) complex to induce engulfment of breast CSCs by immune cells.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Flufenamic Acid/metabolism , Copper/metabolism , Cell Line, Tumor , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Neoplastic Stem Cells
2.
Chemistry ; 30(13): e202303130, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38224207

ABSTRACT

Anilines are core motifs in a variety of important molecules including medicines, materials and agrochemicals. We report a straightforward procedure that allows access to new chemical space of anilines via their para-C-H alkylation. The method utilizes commercially available catalytic H2 O ⋅ B(C6 F5 )3 and is highly selective for para-C-alkylation (over N-alkylation and ortho-C-alkylation) of anilines, with a wide scope in both the aniline substrates and alkene coupling partners. Readily available alkenes are used, and include new classes of alkene for the first time. The mild reaction conditions have allowed the procedure to be applied to the late-stage-functionalization of non-steroidal anti-inflammatory drugs (NSAIDs), including fenamic acids and diclofenac. The formed novel NSAID derivatives display improved anti-inflammatory properties over the parent NSAID structure.


Subject(s)
Alkenes , Aniline Compounds , Alkenes/chemistry , Aniline Compounds/chemistry , Alkylation , Anti-Inflammatory Agents, Non-Steroidal , Catalysis
3.
Angew Chem Int Ed Engl ; 63(6): e202317940, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38063406

ABSTRACT

The cytotoxic and immunogenic-activating properties of a cobalt(III)-cyclam complex bearing the non-steroidal anti-inflammatory drug, flufenamic acid is reported within the context of anti-cancer stem cell (CSC) drug discovery. The cobalt(III)-cyclam complex 1 displays sub-micromolar potency towards breast CSCs grown in monolayers, 24-fold and 31-fold greater than salinomycin (an established anti-breast CSC agent) and cisplatin (an anticancer metallopharmaceutical), respectively. Strikingly, the cobalt(III)-cyclam complex 1 is 69-fold and 50-fold more potent than salinomycin and cisplatin towards three-dimensionally cultured breast CSC mammospheres. Mechanistic studies reveal that 1 induces DNA damage, inhibits cyclooxygenase-2 expression, and prompts caspase-dependent apoptosis. Breast CSCs treated with 1 exhibit damage-associated molecular patterns characteristic of immunogenic cell death and are phagocytosed by macrophages. As far as we are aware, 1 is the first cobalt complex of any oxidation state or geometry to display both cytotoxic and immunogenic-activating effects on breast CSCs.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Heterocyclic Compounds , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cisplatin/pharmacology , Flufenamic Acid/metabolism , Flufenamic Acid/pharmacology , Flufenamic Acid/therapeutic use , Coordination Complexes/metabolism , Cobalt/pharmacology , Cobalt/metabolism , Cell Line, Tumor , Antineoplastic Agents/therapeutic use , Neoplastic Stem Cells
4.
Molecules ; 28(17)2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37687229

ABSTRACT

Cancer stem cells (CSCs) are thought to be partly responsible for metastasis and cancer relapse. Currently, there are no effective therapeutic options that can remove CSCs at clinically safe doses. Here, we report the synthesis, characterisation, and anti-breast CSC properties of a series of copper(I) complexes, comprising of non-steroidal anti-inflammatory drugs (NSAIDs) and triphenylphosphine ligands (1-3). The copper(I) complexes are able to reduce the viability of breast CSCs grown in two- and three-dimensional cultures at micromolar concentrations. The potency of the copper(I) complexes towards breast CSCs was similar to salinomycin (an established anti-breast CSC agent) and cisplatin (a clinically used metallopharmaceutical). Cell-based studies showed that the copper(I) complexes are readily, and similarly, internalised by breast CSCs. The copper(I) complexes significantly increase the intracellular reactive oxygen species (ROS) levels in breast CSCs, and their ROS generation profile with respect to time is dependent on the NSAID component present. The generation of intracellular ROS by the copper(I) complexes could be part of the underlying mechanism by which they evoke breast CSC death. As far as we are aware, this is the first study to explore the anti-breast CSC properties of copper(I) complexes.


Subject(s)
Copper , Neoplasms , Humans , Reactive Oxygen Species , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Neoplastic Stem Cells
5.
Dalton Trans ; 52(28): 9694-9704, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37386843

ABSTRACT

Copper(II)-terpyridine complexes are endowed with the ability to generate reactive oxygen species (ROS) and induce cancer cell death. Here we report the synthesis, characterisation, and anti-breast cancer stem cell (CSC) properties of a series of copper(II)-terpyridine complexes containing aryl sulfonamide groups (1-5). All of the copper(II)-terpyridine complexes adopt distorted square pyramidal geometries and are suitably stable in biologically relevant solutions (PBS and cell culture media). The p-toluene sulfonamide-bearing copper(II)-terpyridine complex 1 is 6-8-fold more potent towards breast CSCs than salinomycin (an established anti-CSC agent) and cisplatin (a metal-based anticancer drug). The copper(II)-terpyridine complex 1 also reduces the formation, size, and viability of three-dimensionally cultured mammospheres, to a similar or better extent than salinomycin and cisplatin. Mechanistic studies show that 1 successfully enters breast CSCs, generates intracellular ROS at short exposure times, partially induces endoplasmic reticulum stress, and triggers apoptosis. To the best of our knowledge, this is the first study to investigate the anti-breast CSC properties of copper(II)-terpyridine complexes.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Cisplatin/metabolism , Copper/pharmacology , Copper/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Neoplastic Stem Cells , Cell Line, Tumor , Neoplasms/metabolism
6.
Chemistry ; 29(45): e202301188, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37249243

ABSTRACT

Mononuclear copper(II)-phenanthroline complexes have been widely investigated as anticancer agents whereas multinuclear copper(II)-phenanthroline complexes are underexplored. Here the synthesis and characterisation of two new binuclear copper(II)-phenanthroline complexes 1 and 2 is reported, comprising of 2,9-dimethyl-1,10-phenanthroline or 2,9-dimethyl-4,7-diphenyl-1,10-phenanthroline, terminal chloride ligands, and bridging chloride or hydroxide ligands. The binuclear copper(II) complex containing 2,9-dimethyl-1,10-phenanthroline 1 displays nanomolar toxicity towards bulk breast cancer cells and breast cancer stem cells (CSCs) grown in monolayers, >50-fold greater than cisplatin (an anticancer metallodrug) and salinomycin (a gold-standard anti-CSC agent). Spectacularly, 1 exhibits >100-fold greater potency toward three-dimensionally cultured mammospheres than cisplatin and salinomycin. Mechanistic studies show that 1 evokes breast CSC apoptosis by elevating intracellular reactive oxygen species levels and damaging genomic DNA (possibly by an oxidative mechanism). To the best of our knowledge, this is the first study to probe the anti-breast CSC properties of binuclear copper(II)-phenanthroline complexes.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Humans , Female , Cisplatin , Copper , Phenanthrolines/pharmacology , Cell Line, Tumor , Coordination Complexes/pharmacology , Breast Neoplasms/drug therapy , Chlorides , Ligands , Antineoplastic Agents/pharmacology , Neoplastic Stem Cells
7.
Molecules ; 28(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36985478

ABSTRACT

Cancer stem cells (CSCs) are linked to tumour relapse and metastasis, the main reason for cancer-related deaths. The application of polymeric nanoparticles as drug delivery systems to target CSCs is relatively unexplored. Here, we report the encapsulation of a CSC-potent copper(II) complex 1 by two compositionally different methoxy poly(ethylene glycol)-b-poly(D,L-lactic-co-glycolic) acid (PEG-PLGA) copolymers. Specifically, we used PEG-PLGA (5000:10,000 Da, 1:1 LA:GA) and PEG-PLGA (5000:10,000 Da, 4:1 LA:GA) polymers to prepare spherical nanoparticle formulations 1:1 NP15 and 4:1 NP15, respectively, both with a 15% feed of 1. The two formulations show distinct biophysical and in vitro properties. For example, (i) 4:1 NP15 displays a slower payload release profile than 1:1 NP15 in physiologically relevant solutions, (ii) 4:1 NP15 exhibits statistically greater potency towards breast CSCs than bulk breast cancer cells grown in monolayers, whereas 1:1 NP15 is equally potent towards breast CSCs and bulk breast cancer cells, and (iii) 4:1 NP15 shows significantly greater potency towards three-dimensionally cultured mammospheres than 1:1 NP15. This study shows that the release profile and anti-breast CSC properties of PEG-PLGA nanoparticle formulations (containing 1) can be perturbed (and possibly controlled) by modifying the proportion of glycolic acid within the PLGA component.


Subject(s)
Breast Neoplasms , Nanoparticles , Humans , Female , Polylactic Acid-Polyglycolic Acid Copolymer , Copper , Polyglycolic Acid , Neoplasm Recurrence, Local , Polyethylene Glycols , Lactic Acid , Breast Neoplasms/drug therapy , Stem Cells , Drug Carriers , Particle Size
8.
Chem Sci ; 14(3): 557-565, 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36741517

ABSTRACT

The anti-breast cancer stem cell (CSC) properties of a series of gold(i) complexes comprising various non-steroidal anti-inflammatory drugs (NSAIDs) and triphenylphosphine 1-8 are reported. The most effective gold(i)-NSAID complex 1, containing indomethacin, exhibits greater potency for breast CSCs than bulk breast cancer cells (up to 80-fold). Furthermore, 1 reduces mammosphere viability to a better extent than a panel of clinically used breast cancer drugs and salinomycin, an established anti-breast CSC agent. Mechanistic studies suggest 1-induced breast CSC death results from breast CSC entry, cytoplasm localisation, an increase in intracellular reactive oxygen species levels, cyclooxygenase-2 downregulation and inhibition, and apoptosis. Remarkably, 1 also significantly inhibits tumour growth in a murine metastatic triple-negative breast cancer model. To the best of our knowledge, 1 is the first gold complex of any geometry or oxidation state to demonstrate anti-breast CSC properties.

9.
ChemMedChem ; 18(4): e202200599, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36533570

ABSTRACT

Here we report the encapsulation of an osteosarcoma stem cell (OSC) potent gallium(III)-diflunisal complex 1 into polymeric nanoparticles, and its delivery into osteosarcoma cells. At the optimum feed (20 %, 1 NP20 ), nanoparticle encapsulation of 1 enhances potency towards bulk osteosarcoma cells and OSCs (cultured in monolayer and three-dimensional systems). Strikingly, the nanoparticle formulation exhibits up to 5645-fold greater potency towards OSCs than frontline anti-osteosarcoma drugs, doxorubicin and cisplatin. The nanoparticle formulation evokes a similar mechanism of action as the payload, which bodes well for future translation. Specifically, the nanoparticle formulation induces nuclear DNA damage, cyclooxygenase-2 downregulation, and caspase-dependent apoptosis. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver an OSC-active metal complex into osteosarcoma cells.


Subject(s)
Bone Neoplasms , Diflunisal , Gallium , Nanoparticles , Osteosarcoma , Humans , Diflunisal/pharmacology , Micelles , Gallium/pharmacology , Cell Line, Tumor , Osteosarcoma/drug therapy , Polymers/pharmacology , Neoplastic Stem Cells
10.
Curr Opin Chem Biol ; 72: 102237, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36542889

ABSTRACT

Cancer stem cells (CSC) are a sub-population of tumours linked to metastasis and relapse. Current chemotherapeutic drug options are ineffective against CSCs at their administered doses. New families of cytotoxic agents, and new, highly specific ways of delivering them to CSCs, are needed to provide durable clinical outcomes. Inorganic compounds have recently emerged as a promising class of anti-CSC agents with clinically relevant potencies. In this short review, we present the very latest efforts (post-2020) on the development of anti-CSC metal complexes. The activities of the metal complexes in monolayer and three-dimensional CSC cultures and animal models is documented. The mechanism of action of the metal complexes with respect to their chemical structures is also highlighted.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Animals , Coordination Complexes/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Chemistry, Inorganic , Antineoplastic Agents/chemistry
11.
Dalton Trans ; 51(47): 18127-18135, 2022 Dec 06.
Article in English | MEDLINE | ID: mdl-36382541

ABSTRACT

The first example of a Pt complex of GANT61, a hedgehog (Hh) pathway inhibitor is reported. Reaction of cis-[Pt(II)Cl2(dmso)2] with one equivalent of 4-pyridine carboxaldehyde (4-PCA, control ligand) or one equivalent of GANT61 (Hh pathway inhibitor) in acetone at rt for 30 minutes afforded trans-[Pt(II)Cl2(dmso)(4-PCA)] (1) and trans-[Pt(II)Cl2(dmso)(GANT61)] (2) respectively, where 4-PCA and GANT61 are N-donor ligands. The structures of 1 and 2 were fully characterised by elemental analysis, 1H NMR, 13C NMR and IR spectroscopy and X-ray crystallography. 1 and 2 undergo isomerisation from trans- to cis-in solution and therefore the biological activity of 2 is also associated with the cis-configuration. The in vitro cytotoxicity data show that 2 is a potent inhibitor of the growth of breast CSC-depleted HMLER and breast CSC-enriched HMLER-shEcad cells. Furthermore 2 markedly reduced the size and viability and significantly reduced the number of CSC-enriched HMLER-shEcad mammospheres formed. 2 also induced apoptosis with low micromolar IC50 values against two triple negative breast cancer lines, MDA-MB-231 (MDA231) and BT549. 2, which possesses the Hh pathway inhibitor GANT61 as an N donor ligand exhibits far superior anti-CSC activity including in the CSC-enriched mammosphere model and activity against TNBC cells as compared to its control analogue, the trans-Pt(II) 4-PCA complex 1. The trans-Pt GANT61 complex 2 has also been shown to cause DNA damage and inhibit the Hh pathway at the level of GLI.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Hedgehog Proteins , Ligands , Neoplastic Stem Cells
12.
Chembiochem ; 23(24): e202200532, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36281941

ABSTRACT

We report the synthesis, characterisation, and anti-osteosarcoma properties of a gallium(III) complex (1) comprising of two 1,10-phenanthroline ligands and salicylate, a non-steroidal anti-inflammatory drug. The gallium(III) complex 1 displays micromolar potency towards bulk osteosarcoma cells and osteosarcoma stem cells (OSCs). Notably, the gallium(III) complex 1 exhibits significantly higher toxicity towards OSCs grown in monolayer and three-dimensional cultures than cisplatin, a frontline anti-osteosarcoma drug. Nuclei isolation and immunoblotting studies show that the gallium(III) complex 1 enters osteosarcoma cell nuclei and induces DNA damage. Flow cytometry and cytotoxicity studies (in the presence of prostaglandin E2) indicate that the gallium(III) complex 1 downregulates cyclooxygenase-2 (COX-2) expression and kills osteosarcoma cells in a COX-2-dependent manner. Further, the mode of osteosarcoma cell death evoked by the gallium(III) complex 1 is characterised as caspase-dependent apoptosis.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Gallium , Osteosarcoma , Humans , Phenanthrolines/pharmacology , Gallium/pharmacology , Gallium/therapeutic use , Salicylates/pharmacology , Salicylates/therapeutic use , Cyclooxygenase 2/metabolism , Cell Line, Tumor , Osteosarcoma/drug therapy , Apoptosis , Stem Cells/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
13.
Molecules ; 27(10)2022 May 19.
Article in English | MEDLINE | ID: mdl-35630754

ABSTRACT

Apoptosis resistance is inherent to stem cell-like populations within tumours and is one of the major reasons for chemotherapy failures in the clinic. Necroptosis is a non-apoptotic mode of programmed cell death that could help bypass apoptosis resistance. Here we report the synthesis, characterisation, biophysical properties, and anti-osteosarcoma stem cell (OSC) properties of a new nickel(II) complex bearing 3,4,7,8-tetramethyl-1,10-phenanthroline and two flufenamic acid moieties, 1. The nickel(II) complex 1 is stable in both DMSO and cell media. The nickel(II) complex 1 kills bulk osteosarcoma cells and OSCs grown in monolayer cultures and osteospheres grown in three-dimensional cultures within the micromolar range. Remarkably, 1 exhibits higher potency towards osteospheres than the metal-based drugs used in current osteosarcoma treatment regimens, cisplatin and carboplatin, and an established anti-cancer stem cell agent, salinomycin (up to 7.7-fold). Cytotoxicity studies in the presence of prostaglandin E2 suggest that 1 kills OSCs in a cyclooxygenase-2 (COX-2) dependent manner. Furthermore, the potency of 1 towards OSCs decreased significantly upon co-treatment with necrostatin-1 or dabrafenib, well-known necroptosis inhibitors, implying that 1 induces necroptosis in OSCs. To the best of our knowledge, 1 is the first compound to implicate both COX-2 and necroptosis in its mechanism of action in OSCs.


Subject(s)
Bone Neoplasms , Coordination Complexes , Osteosarcoma , Bone Neoplasms/pathology , Cell Line, Tumor , Coordination Complexes/pharmacology , Cyclooxygenase 2/metabolism , Flufenamic Acid , Humans , Neoplastic Stem Cells/metabolism , Nickel/metabolism , Nickel/pharmacology , Osteosarcoma/pathology
14.
RSC Adv ; 12(9): 5290-5299, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35425564

ABSTRACT

The major cause for cancer related deaths worldwide is tumour relapse and metastasis, both of which have been heavily linked to the existence of cancer stem cells (CSCs). CSCs are able to escape current treatment regimens, reform tumours, and promote their spread to secondary sites. Recently, our research group reported the first metal-based agent 1 (a copper(ii) compound ligated by a bidentate 4,7-diphenyl-1,10-phenanthroline and a tridentate Schiff base ligand) to potently kill CSCs via cytotoxic and immunogenic mechanisms. Here we show that encapsulation of 1 by polymeric nanoparticles at the appropriate feed (10%, 1 NP10) enhances CSC uptake and improves potency towards bulk cancer cells and CSCs (grown in monolayer and three-dimensional cultures). The nanoparticle formulation triggers a similar cellular response to the payload, which bodes well for further translation. Specifically, the nanoparticle formulation elevates intracellular reactive oxygen species levels, induces ER stress, and evokes damage-associated molecular patterns consistent with immunogenic cell death. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver immunogenic metal complexes into CSCs.

15.
Adv Sci (Weinh) ; 9(13): e2105506, 2022 05.
Article in English | MEDLINE | ID: mdl-35246961

ABSTRACT

Membrane-lytic peptides offer broad synthetic flexibilities and design potential to the arsenal of anticancer therapeutics, which can be limited by cytotoxicity to noncancerous cells and induction of drug resistance via stress-induced mutagenesis. Despite continued research efforts on membrane-perforating peptides for antimicrobial applications, success in anticancer peptide therapeutics remains elusive given the muted distinction between cancerous and normal cell membranes and the challenge of peptide degradation and neutralization upon intravenous delivery. Using triple-negative breast cancer as a model, the authors report the development of a new class of anticancer peptides. Through function-conserving mutations, the authors achieved cancer cell selective membrane perforation, with leads exhibiting a 200-fold selectivity over non-cancerogenic cells and superior cytotoxicity over doxorubicin against breast cancer tumorspheres. Upon continuous exposure to the anticancer peptides at growth-arresting concentrations, cancer cells do not exhibit resistance phenotype, frequently observed under chemotherapeutic treatment. The authors further demonstrate efficient encapsulation of the anticancer peptides in 20 nm polymeric nanocarriers, which possess high tolerability and lead to effective tumor growth inhibition in a mouse model of MDA-MB-231 triple-negative breast cancer. This work demonstrates a multidisciplinary approach for enabling translationally relevant membrane-lytic peptides in oncology, opening up a vast chemical repertoire to the arms race against cancer.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Humans , Mice , Peptides , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
16.
Dalton Trans ; 51(15): 5904-5912, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35348171

ABSTRACT

Copper(II) coordination compounds have been investigated for their anticancer properties for decades, however, none have reached advanced human clinical trials. The poor translation of copper(II) complexes from in vitro studies to (pre)clinical studies can be attributed to their limited efficacy in animal models, which is largely associated with copper leaching and speciation (in biological fluids). Here we report a biologically stable copper(II) complex based on the active site of Type I Cu electron transport proteins. The copper(II) complex 1 comprises of dithiacyclam (with soft and hard donor atoms) and two diclofenac units, a nonsteriodial anti-inflammatory drug (NSAID). Extensive biophysical and electrochemical studies show that the solid state structure of 1 is preserved in solution and that it can access both copper(I) and copper(II) oxidation states without leaching copper or undergoing speciation (in the presence of a cellular reductant). Cell studies show that 1 kills bulk breast cancer cells and highly resistant breast cancer stem cells (CSCs) at micromolar concentrations, and is significantly less toxic towards a panel of non-cancerous cells. Clinically relevant spheroid studies show that 1 is able to inhibit breast CSC-enriched mammosphere formation to a similar extent as salinomycin, a gold standard anti-CSC agent. Mechanistic studies show that 1 evokes breast CSC death by elevating intracellular reactive oxygen species (ROS) and inhibiting cyclooxygenase-2 (COX-2) activity. The former leads to the activation of stress pathways (JNK and p38), which culminates in caspase-dependent apoptosis. This study reinforces the therapeutic potential of copper(II)-NSAID complexes and provides a bioinspired route to develop stable, ROS-generating copper-based anti-CSC drug candidates.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Coordination Complexes/chemistry , Copper/chemistry , Neoplastic Stem Cells , Oxidation-Reduction , Reactive Oxygen Species/metabolism
17.
Chemistry ; 27(55): 13846-13854, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34269487

ABSTRACT

We report the anti-osteosarcoma stem cell (OSC) properties of a series of gallium(III)-polypyridyl complexes (5-7) containing diflunisal, a non-steroidal anti-inflammatory drug. The most effective complex within the series, 6 (containing 3,4,7,8-tetramethyl-1,10-phenanthroline), displayed similar potency towards bulk osteosarcoma cells and OSCs, in the nanomolar range. Remarkably, 6 exhibited significantly higher monolayer and sarcosphere OSC potency (up to three orders of magnitude) than clinically approved drugs used in frontline (cisplatin and doxorubicin) and secondary (etoposide, ifosfamide, and carboplatin) osteosarcoma treatments. Mechanistic studies show that 6 downregulates cyclooxygenase-2 (COX-2) and kills osteosarcoma cells in a COX-2 dependent manner. Furthermore, 6 induces genomic DNA damage and caspase-dependent apoptosis. To the best of our knowledge, 6 is the first metal complex to kill osteosarcoma cells by simultaneously inhibiting COX-2 and damaging nuclear DNA.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Diflunisal , Gallium , Osteosarcoma , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Cell Line, Tumor , Diflunisal/therapeutic use , Humans , Neoplastic Stem Cells , Osteosarcoma/drug therapy
18.
Dalton Trans ; 50(17): 5779-5783, 2021 May 07.
Article in English | MEDLINE | ID: mdl-33890607

ABSTRACT

A cancer stem cell (CSC) active, solution stable, silver(i) polymeric complex bearing a dithiacyclam ligand is reported. The complex displays similar potency towards CSCs to salinomycin in monolayer and three-dimensional cultures. Mechanistic studies suggest CSC death results from cytosol entry, an increase in intracellular reactive oxygen species, and caspase-dependent apoptosis.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Neoplastic Stem Cells/drug effects , Polymers/chemistry , Silver/chemistry , Cell Line, Tumor , Humans , Reactive Oxygen Species/metabolism
19.
Angew Chem Int Ed Engl ; 60(12): 6704-6709, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33274606

ABSTRACT

We report the anti-breast cancer stem cell (CSC) properties of a series of Group 10-bis(azadiphosphine) complexes 1-3 under exclusively three-dimensional cell culture conditions. The breast CSC mammosphere potency of 1-3 is dependent on the Group 10 metal present, increasing in the following order: 1 (nickel complex) <2 (palladium complex) <3 (platinum complex). Notably, 3 reduces the formation and size of mammospheres to a greater extent than salinomycin, an established CSC-active compound, or any reported anti-CSC metal complex tested under similar conditions. Mechanistic studies suggest that the most effective complexes 2 and 3 readily penetrate CSC mammospheres, enter CSC nuclei, induce genomic DNA damage, and trigger caspase-dependent apoptosis. To the best of our knowledge, this is the first study to systematically probe the anti-CSC activity of a series of structurally related Group 10 complexes and to be conducted entirely using three-dimensional CSC culture conditions.


Subject(s)
Antineoplastic Agents/pharmacology , Aza Compounds/pharmacology , Breast Neoplasms/drug therapy , Coordination Complexes/pharmacology , Neoplastic Stem Cells/drug effects , Phosphines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aza Compounds/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , DNA Damage , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Particle Size , Phosphines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
20.
Chembiochem ; 21(24): 3618-3624, 2020 12 11.
Article in English | MEDLINE | ID: mdl-32776422

ABSTRACT

Immunogenic cell death (ICD) offers a method of stimulating the immune system to attack and remove cancer cells. We report a copper(II) complex containing a Schiff base ligand and a polypyridyl ligand, 4, capable of inducing ICD in breast cancer stem cells (CSCs). Complex 4 kills both bulk breast cancer cells and breast CSCs at sub-micromolar concentrations. Notably, 4 exhibits greater potency (one order of magnitude) towards breast CSCs than salinomycin (an established breast CSC-potent agent) and cisplatin (a clinically approved anticancer drug). Epithelial spheroid studies show that 4 is able to selectively inhibit breast CSC-enriched HMLER-shEcad spheroid formation and viability over non-tumorigenic breast MCF10 A spheroids. Mechanistic studies show that 4 operates as a Type II ICD inducer. Specifically, 4 readily enters the endoplasmic reticulum (ER) of breast CSCs, elevates intracellular reactive oxygen species (ROS) levels, induces ER stress, evokes damage-associated molecular patterns (DAMPs), and promotes breast CSC phagocytosis by macrophages. As far as we are aware, 4 is the first metal complex to induce ICD in breast CSCs and promote their engulfment by immune cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coordination Complexes/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemistry , Copper/chemistry , Copper/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Humans , Immunogenic Cell Death/drug effects , Immunogenic Cell Death/immunology , Ligands , Models, Molecular , Molecular Structure , Reactive Oxygen Species/immunology , Schiff Bases/chemistry , Schiff Bases/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...