Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Genes Immun ; 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38135730
2.
Nat Biotechnol ; 41(7): 980-992, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36593401

ABSTRACT

Identification of CD8+ T cell epitopes is critical for the development of immunotherapeutics. Existing methods for major histocompatibility complex class I (MHC class I) ligand discovery are time intensive, specialized and unable to interrogate specific proteins on a large scale. Here, we present EpiScan, which uses surface MHC class I levels as a readout for whether a genetically encoded peptide is an MHC class I ligand. Predetermined starting pools composed of >100,000 peptides can be designed using oligonucleotide synthesis, permitting large-scale MHC class I screening. We exploit this programmability of EpiScan to uncover an unappreciated role for cysteine that increases the number of predicted ligands by 9-21%, reveal affinity hierarchies by analysis of biased anchor peptide libraries and screen viral proteomes for MHC class I ligands. Using these data, we generate and iteratively refine peptide binding predictions to create EpiScan Predictor. EpiScan Predictor performs comparably to other state-of-the-art MHC class I peptide binding prediction algorithms without suffering from underrepresentation of cysteine-containing peptides. Thus, targeted immunopeptidomics using EpiScan will accelerate CD8+ T cell epitope discovery toward the goal of individual-specific immunotherapeutics.


Subject(s)
Cysteine , Histocompatibility Antigens Class I , Ligands , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Peptides/metabolism , Epitopes, T-Lymphocyte/genetics , Protein Binding
4.
Proc Natl Acad Sci U S A ; 117(8): 4053-4060, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32041867

ABSTRACT

Small molecules can affect many cellular processes. The disambiguation of these effects to identify the causative mechanisms of cell death is extremely challenging. This challenge impacts both clinical development and the interpretation of chemical genetic experiments. CX-5461 was developed as a selective RNA polymerase I inhibitor, but recent evidence suggests that it may cause DNA damage and induce G-quadraplex formation. Here we use three complimentary data mining modalities alongside biochemical and cell biological assays to show that CX-5461 exerts its primary cytotoxic activity through topoisomerase II poisoning. We then show that acquired resistance to CX-5461 in previously sensitive lymphoma cells confers collateral resistance to the topoisomerase II poison doxorubicin. Doxorubicin is already a frontline chemotherapy in a variety of hematopoietic malignancies, and CX-5461 is being tested in relapse/refractory hematopoietic tumors. Our data suggest that the mechanism of cell death induced by CX-5461 is critical for rational clinical development in these patients. Moreover, CX-5461 usage as a specific chemical genetic probe of RNA polymerase I function is challenging to interpret. Our multimodal data-driven approach is a useful way to detangle the intended and unintended mechanisms of drug action across diverse essential cellular processes.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Cell Survival/drug effects , Naphthyridines/pharmacology , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Cell Line, Tumor , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Interference , Sensitivity and Specificity
5.
Nat Commun ; 9(1): 628, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416025

ABSTRACT

The original version of this Article contained an error in the spelling of the author Yingjie Yu, which was incorrectly given as Yu Yingjie. Furthermore, in Figure 3a, the labels 'MD | p < 0.05' incorrectly read 'MD | p > 0.05'. These errors have now been corrected in both the PDF and HTML versions of the Article.

6.
Nat Commun ; 8(1): 2166, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29255160

ABSTRACT

Advanced-stage epithelial ovarian cancers are amongst the most difficult to treat tumors and have proven to be refractory to most cytotoxic, molecularly targeted, or immunotherapeutic approaches. Here, we report that nanoparticle-drug conjugates (NDCs) of monomethyl auristatin E (MMAE) significantly increase loading on a per-vehicle basis as compared to antibody-drug conjugates (ADCs). Their intraperitoneal administration enabled triggered release of the active MMAE toxin to inhibit tumor growth and to extend animal survival to >90 days in a cell-line xenograft model of disseminated ovarian cancer. In a patient-derived xenograft model of advanced-stage and platinum-resistant ovarian cancer, an MMAE-based NDC doubled the duration of tumor growth inhibition as compared to cisplatin. NDCs of highly potent toxins thus introduce a translatable platform that may be exploited to maximize the safety and efficacy of cytotoxic chemotherapies, combining the best features of ADCs with those of nanoparticle-based therapeutics.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Immunoconjugates/pharmacology , Nanoparticles/chemistry , Ovarian Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Survival Analysis , Treatment Outcome
7.
Cancer Res ; 77(19): 5272-5286, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28775166

ABSTRACT

Aneuploidy, a hallmark of cancer cells, poses an appealing opportunity for cancer treatment and prevention strategies. Using a cell-based screen to identify small molecules that could selectively kill aneuploid cells, we identified the compound N-[2-hydroxy-1-(4-morpholinylmethyl)-2-phenylethyl]-decanamide monohydrochloride (DL-PDMP), an antagonist of UDP-glucose ceramide glucosyltransferase. DL-PDMP selectively inhibited proliferation of aneuploid primary mouse embryonic fibroblasts and aneuploid colorectal cancer cells. Its selective cytotoxic effects were based on further accentuating the elevated levels of ceramide, which characterize aneuploid cells, leading to increased apoptosis. We observed that DL-PDMP could also enhance the cytotoxic effects of paclitaxel, a standard-of-care chemotherapeutic agent that causes aneuploidy, in human colon cancer and mouse lymphoma cells. Our results offer pharmacologic evidence that the aneuploid state in cancer cells can be targeted selectively for therapeutic purposes, or for reducing the toxicity of taxane-based drug regimens. Cancer Res; 77(19); 5272-86. ©2017 AACR.


Subject(s)
Aneuploidy , Colorectal Neoplasms/pathology , Embryo, Mammalian/cytology , Fibroblasts/cytology , Homeostasis , Lymphoma/pathology , Sphingolipids/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Ceramides/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Synergism , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Glucosyltransferases/metabolism , Humans , Lymphoma/drug therapy , Lymphoma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Morpholines/pharmacology , Sphingosine N-Acyltransferase/metabolism , Xenograft Model Antitumor Assays
8.
Chemistry ; 23(40): 9674-9682, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28556445

ABSTRACT

The cytotoxic properties of a series of nickel(II)-dithiocarbamate phenanthroline complexes is reported. The complexes 1-6 kill bulk cancer cells and cancer stem cells (CSCs) with micromolar potency. Two of the complexes, 2 and 6, kill twice as many breast cancer stem cell (CSC)-enriched HMLER-shEcad cells as compared to breast CSC-depleted HMLER cells. Complex 2 inhibits mammosphere formation to a similar extent as salinomycin (a CSC-specific toxin). Detailed mechanistic studies suggest that 2 induces CSC death by necroptosis, a programmed form of necrosis. Specifically, 2 triggers MLKL phosphorylation, oligomerization, and translocation to the cell membrane. Additionally, 2 induces necrosome-mediated propidium iodide (PI) uptake and mitochondrial membrane depolarisation, as well as morphological changes consistent with necroptotosis. Strikingly, 2 does not evoke necroptosis by intracellular reactive oxygen species (ROS) production or poly(ADP) ribose polymerase (PARP-1) activation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Coordination Complexes/chemical synthesis , Neoplastic Stem Cells/drug effects , Nickel/chemistry , Phenanthrolines/chemical synthesis , Thiocarbamates/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Coordination Complexes/pharmacology , Humans , Necrosis , Neoplastic Stem Cells/pathology , Phenanthrolines/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Reactive Oxygen Species/metabolism , Thiocarbamates/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors
9.
Nat Med ; 23(4): 461-471, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28263311

ABSTRACT

Cisplatin and its platinum analogs, carboplatin and oxaliplatin, are some of the most widely used cancer chemotherapeutics. Although cisplatin and carboplatin are used primarily in germ cell, breast and lung malignancies, oxaliplatin is instead used almost exclusively to treat colorectal and other gastrointestinal cancers. Here we utilize a unique, multi-platform genetic approach to study the mechanism of action of these clinically established platinum anti-cancer agents, as well as more recently developed cisplatin analogs. We show that oxaliplatin, unlike cisplatin and carboplatin, does not kill cells through the DNA-damage response. Rather, oxaliplatin kills cells by inducing ribosome biogenesis stress. This difference in drug mechanism explains the distinct clinical implementation of oxaliplatin relative to cisplatin, and it might enable mechanistically informed selection of distinct platinum drugs for distinct malignancies. These data highlight the functional diversity of core components of front-line cancer therapy and the potential benefits of applying a mechanism-based rationale to the use of our current arsenal of anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Cisplatin/pharmacology , Neoplasms , Organelle Biogenesis , Organoplatinum Compounds/pharmacology , Ribosomes/drug effects , Animals , Blotting, Western , Cell Line, Tumor , DNA Damage/drug effects , Humans , Mice , Oxaliplatin , Phenanthridines/pharmacology , Platinum Compounds/pharmacology , Principal Component Analysis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Stress, Physiological , Xenograft Model Antitumor Assays
10.
Inorganica Chim Acta ; 452: 125-129, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27818526

ABSTRACT

The cellular response evoked by a hexanuclear platinum complex, Pt6L4 (1), is reported. Compound 1, a 3-nm octahedral cage formed by self-assembly of six Pt(II) centers and four 2,4,6-tris(4-pyridyl)-1,3,5-triazine ligands (L), exhibits promising in vitro potency against a panel of human cancer cell lines. Unlike classical platinum-based anticancer agents, 1 interacts with DNA in a non-covalent, intercalative manner and promotes DNA condensation. In cancer cells, 1 induces DNA damage, upregulates p53, its phosphorylated form phospho-p53 and its downstream effector, p21, as well as both apoptosis and senescence.

11.
Dalton Trans ; 45(44): 17867-17873, 2016 Nov 28.
Article in English | MEDLINE | ID: mdl-27774561

ABSTRACT

We report the cancer stem cell (CSC) potency of a novel series of copper(ii)-phenanthroline complexes bearing nonsteriodial anti-inflammatory drugs: naproxen, tolfenamic acid, and indomethacin (2a-3c). Two of the complexes, 2a and 3c, kill breast CSC-enriched HMLER-shEcad cells (grown in both monolayer and three-dimensional cell cultures) to a significantly better extent than salinomycin, a well-established CSC toxin. The most potent complex in the series, 3c induces its cytotoxic effect by generating intracellular reactive oxygen species (ROS) and inhibiting cyclooxgenase-2 (COX-2) activity. Encapsulation of 3c using biodegradable methoxy poly(ethylene glycol)-b-poly(d,l-lactic-co-glycolic) acid (PEG-PLGA) copolymers at the appropriate feed (5%, 3c NP5) enhances breast CSC uptake and reduces overall toxicity. The nanoparticle formulation, 3c NP5 selectively kills breast CSCs over bulk breast cancer cells, and evokes a similar cellular response to the payload, 3c. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver CSC-potent metal complexes into CSCs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coordination Complexes/pharmacology , Copper/pharmacology , Neoplastic Stem Cells/drug effects , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Copper/administration & dosage , Copper/chemistry , Drug Carriers/chemistry , Female , Humans , Nanoparticles/chemistry , Neoplastic Stem Cells/pathology , Polyesters/chemistry , Polyethylene Glycols/chemistry
12.
J Am Chem Soc ; 138(38): 12494-501, 2016 09 28.
Article in English | MEDLINE | ID: mdl-27626288

ABSTRACT

Single-nanoparticle (NP) combination chemotherapeutics are quickly emerging as attractive alternatives to traditional chemotherapy due to their ability to increase drug solubility, reduce off-target toxicity, enhance blood circulation lifetime, and increase the amount of drug delivered to tumors. In the case of NP-bound drugs, that is, NP-prodrugs, the current standard of practice is to assume that the subcellular mechanism of action for each drug released from the NP mirrors that of the unbound, free-drug. Here, we use an RNAi signature assay for the first time to examine the mechanism of action of multidrug-conjugated NP prodrugs relative to their small molecule prodrugs and native drug mechanisms of action. Additionally, the effective additive contribution of three different drugs in a single-NP platform is characterized. The results indicate that some platinum(IV) cisplatin prodrugs, although cytotoxic, may not have the expected mechanism of action for cisplatin. This insight was utilized to develop a novel platinum(IV) oxaliplatin prodrug and incorporate it into a three-drug-conjugated NP, where each drug's mechanism of action is preserved, to treat tumor-bearing mice with otherwise lethal levels of chemotherapy.


Subject(s)
Camptothecin/therapeutic use , Doxorubicin/therapeutic use , Nanoparticles/chemistry , Organoplatinum Compounds/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Camptothecin/administration & dosage , Carcinoma/drug therapy , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Delivery Systems , Mice , Molecular Structure , Neoplasms, Experimental/drug therapy , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prodrugs/administration & dosage , RNA Interference
13.
Chembiochem ; 17(18): 1713-8, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27377813

ABSTRACT

We report the potency against cancer stem cells (CSCs) of a new cobalt(III)-cyclam complex (1) that bears the nonsteroidal anti-inflammatory drug, naproxen. The complex displays selective potency for breast CSC-enriched HMLER-shEcad cells over breast CSC-depleted HMLER cells. Additionally, it inhibited the formation of three-dimensional tumour-like mammospheres, and reduced their viability to a greater extent than clinically used breast cancer drugs (vinorelbine, cisplatin and paclitaxel). The anti-mammosphere potency of 1 was enhanced under hypoxia-mimicking conditions. Detailed mechanistic studies revealed that DNA damage and cyclooxygenase-2 (COX-2) inhibition contribute to the cytotoxic mechanism of 1. To the best of our knowledge, 1 is the first cobalt-containing compound to show selective potency for CSCs over bulk cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cobalt/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Naproxen/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Organometallic Compounds/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cobalt/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , DNA Damage/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Conformation , Naproxen/chemistry , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 113(27): E3892-900, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27325776

ABSTRACT

Targeted transcriptional regulation is a powerful tool to study genetic mediators of cellular behavior. Here, we show that catalytically dead Cas9 (dCas9) targeted to genomic regions upstream or downstream of the transcription start site allows for specific and sustainable gene-expression level alterations in tumor cells in vitro and in syngeneic immune-competent mouse models. We used this approach for a high-coverage pooled gene-activation screen in vivo and discovered previously unidentified modulators of tumor growth and therapeutic response. Moreover, by using dCas9 linked to an activation domain, we can either enhance or suppress target gene expression simply by changing the genetic location of dCas9 binding relative to the transcription start site. We demonstrate that these directed changes in gene-transcription levels occur with minimal off-target effects. Our findings highlight the use of dCas9-mediated transcriptional regulation as a versatile tool to reproducibly interrogate tumor phenotypes in vivo.


Subject(s)
Bacterial Proteins/genetics , Endonucleases/genetics , Gene Expression Regulation, Leukemic , Genetic Techniques , Leukemia, Experimental , Animals , CRISPR-Associated Protein 9 , DNA Damage , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Mice , Sequence Analysis, RNA , Temozolomide , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
16.
Angew Chem Int Ed Engl ; 55(8): 2845-50, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26806362

ABSTRACT

The breast cancer stem cell (CSC) potency of a series of copper(II)-phenanthroline complexes containing the nonsteroidal anti-inflammatory drug (NSAID), indomethacin, is reported. The most effective copper(II) complex in this series, 4, selectivity kills breast CSC-enriched HMLER-shEcad cells over breast CSC-depleted HMLER cells. Furthermore, 4 reduces the formation, size, and viability of mammospheres, to a greater extent than salinomycin, a potassium ionophore known to selectively inhibit CSCs. Mechanistic studies revealed that the CSC-specificity observed for 4 arises from its ability to generate intracellular reactive oxygen species (ROS) and inhibit cyclooxygenase-2 (COX-2), an enzyme that is overexpressed in breast CSCs. The former induces DNA damage, activates JNK and p38 pathways, and leads to apoptosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Breast Neoplasms/pathology , Copper/chemistry , Neoplastic Stem Cells/pathology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Female , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism
17.
J Am Chem Soc ; 137(47): 14854-7, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26561720

ABSTRACT

Expression of indoleamine-2,3-dioxygenase (IDO), an immunosuppressive enzyme in human tumors, leads to immune evasion and tumor tolerance. IDO is therefore a tumor immunotherapeutic target, and several IDO inhibitors are currently undergoing clinical trials. IDO inhibitors can enhance the efficacy of common cancer chemotherapeutics. Here we investigate Pt(IV)-(D)-1-methyltryptophan conjugates 1 and 2 for combined immunomodulation and DNA cross-link-triggered apoptosis for cancer "immuno-chemotherapy". Compound 2 effectively kills hormone-dependent, cisplatin-resistant human ovarian cancer cells, inhibiting IDO by transcriptional deregulation of the autocrine-signaling loop IDO-AHR-IL6, which blocks kynurenine production and promotes T-cell proliferation. Additionally, 1 and 2 display low toxicity in mice and are stable in blood. To our knowledge, this construct is the first Pt drug candidate with immune checkpoint blockade properties.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Ovarian Neoplasms/therapy , Female , Humans , Ovarian Neoplasms/drug therapy
18.
J Am Chem Soc ; 137(8): 2967-74, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25698398

ABSTRACT

Rhenium(V) oxo complexes of general formula [ReO(OMe)(N^N)Cl2], where N^N = 4,7-diphenyl-1,10-phenanthroline, 1, or 3,4,7,8-tetramethyl-1,10-phenanthroline, 2, effectively kill cancer cells by triggering necroptosis, a non-apoptotic form of cell death. Both complexes evoke necrosome (RIP1-RIP3)-dependent intracellular reactive oxygen species (ROS) production and propidium iodide uptake. The complexes also induce mitochondrial membrane potential depletion, a possible downstream effect of ROS production. Apparently, 1 and 2 are the first rhenium complexes to evoke cellular events consistent with programmed necrosis in cancer cells. Furthermore, 1 and 2 display low acute toxicity in C57BL/6 mice and reasonable stability in fresh human blood.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Phenanthrolines/chemistry , Rhenium/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Cycle/drug effects , Cell Line, Tumor , Drug Stability , Humans , Mice , Models, Molecular , Molecular Conformation , Necrosis/chemically induced , Organometallic Compounds/chemical synthesis , Organometallic Compounds/toxicity , Receptor, PAR-1/metabolism , Tumor Suppressor Protein p53/metabolism
19.
J Am Chem Soc ; 136(41): 14413-6, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25247635

ABSTRACT

The effect of a newly developed osmium(VI) nitrido complex, 1, on breast cancer stem cells (CSCs) is reported. The complex displays selective toxicity for HMLER breast cancer cells enriched with CD44-positive, CSC-like cells over the same cells having reduced CSC character. Remarkably, 1 also reduces the proportion of CSCs within a heterogeneous breast cancer cell population and irreversibly inhibits the formation of free-floating mammospheres to an extent similar to that of salinomycin, a natural product that targets CSCs. Detailed mechanistic studies reveal that in breast cancer cells 1 induces DNA damage and endoplasmic reticulum stress, the latter being responsible for the CSC selectivity. The anti-CSC properties of 1 provide a strong impetus for the development of new metal-based compounds to target CSCs and to treat chemotherapy-resistant and relapsed tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coordination Complexes/pharmacology , Neoplastic Stem Cells/drug effects , Nitrogen Compounds/chemistry , Osmium/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Conformation , Neoplastic Stem Cells/pathology , Structure-Activity Relationship
20.
J Am Chem Soc ; 135(38): 14060-3, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24041161

ABSTRACT

The cellular response evoked by antiproliferating osmium(VI) nitrido compounds of general formula OsN(N^N)Cl3 (N^N = 2,2'-bipyridine 1, 1,10-phenanthroline 2, 3,4,7,8-tetramethyl-1,10-phenanthroline 3, or 4,7-diphenyl-1,10-phenanthroline 4) can be tuned by subtle ligand modifications. Complex 2 induces DNA damage, resulting in activation of the p53 pathway, cell cycle arrest at the G2/M phase, and caspase-dependent apoptotic cell death. In contrast, 4 evokes endoplasmic reticulum (ER) stress leading to the upregulation of proteins of the unfolded protein response pathway, increase in ER size, and p53-independent apoptotic cell death. To the best of our knowledge, 4 is the first osmium compound to induce ER stress in cancer cells.


Subject(s)
2,2'-Dipyridyl/chemistry , Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemical synthesis , Osmium , Phenanthrolines/chemistry , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , DNA Damage , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Stress , G2 Phase Cell Cycle Checkpoints , Humans , Ligands , M Phase Cell Cycle Checkpoints , Signal Transduction , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...