Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(24): e2404668121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833473

ABSTRACT

Developing anticancer drugs with low side effects is an ongoing challenge. Immunogenic cell death (ICD) has received extensive attention as a potential synergistic modality for cancer immunotherapy. However, only a limited set of drugs or treatment modalities can trigger an ICD response and none of them have cytotoxic selectivity. This provides an incentive to explore strategies that might provide more effective ICD inducers free of adverse side effects. Here, we report a metal-based complex (Cu-1) that disrupts cellular redox homeostasis and effectively stimulates an antitumor immune response with high cytotoxic specificity. Upon entering tumor cells, this Cu(II) complex enhances the production of intracellular radical oxidative species while concurrently depleting glutathione (GSH). As the result of heightening cellular oxidative stress, Cu-1 gives rise to a relatively high cytotoxicity to cancer cells, whereas normal cells with low levels of GSH are relatively unaffected. The present Cu(II) complex initiates a potent ferroptosis-dependent ICD response and effectively inhibits in vivo tumor growth in an animal model (c57BL/6 mice challenged with colorectal cancer). This study presents a strategy to develop metal-based drugs that could synergistically potentiate cytotoxic selectivity and promote apoptosis-independent ICD responses through perturbations in redox homeostasis.


Subject(s)
Copper , Glutathione , Homeostasis , Oxidation-Reduction , Animals , Mice , Humans , Glutathione/metabolism , Mice, Inbred C57BL , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Oxidative Stress/drug effects , Drug Synergism , Immunogenic Cell Death/drug effects , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism
2.
J Med Chem ; 67(8): 6738-6748, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38526421

ABSTRACT

The development and optimization of metal-based anticancer drugs with novel cytotoxic mechanisms have emerged as key strategies to overcome chemotherapeutic resistance and side effects. Agents that simultaneously induce ferroptosis and autophagic death have received extensive attention as potential modalities for cancer therapy. However, only a limited set of drugs or treatment modalities can synergistically induce ferroptosis and autophagic tumor cell death. In this work, we designed and synthesized four new cycloplatinated (II) complexes harboring an isoquinoline alkaloid C∧N ligand. On screening the in vitro activity of these agents, we found that Pt-3 exhibited greater selectivity of cytotoxicity, decreased resistance factors, and improved anticancer activity compared to cisplatin. Furthermore, Pt-3, which we demonstrate can initiate potent ferritinophagy-dependent ferroptosis, exhibits less toxic and better therapeutic activity than cisplatin in vivo. Our results identify Pt-3 as a promising candidate or paradigm for further drug development in cancer treatment.


Subject(s)
Antineoplastic Agents , Ferroptosis , Isoquinolines , Triple Negative Breast Neoplasms , Ferroptosis/drug effects , Humans , Isoquinolines/pharmacology , Isoquinolines/chemistry , Isoquinolines/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Animals , Female , Cell Line, Tumor , Ferritins/metabolism , Autophagy/drug effects , Mice , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Alkaloids/pharmacology , Alkaloids/chemistry , Alkaloids/chemical synthesis , Structure-Activity Relationship , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Mice, Nude
3.
Anal Chim Acta ; 1284: 341993, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37996165

ABSTRACT

It is a great challenge to develop an efficient and rapid method to detect of biomarkers of cardiovascular disease. In this research, a differential pulse voltammetry (DPV)-based ultrasensitive immunosensor for the detection of plasma Latexin (LXN) has been established. With the aim to increase the surface area of the bare glassy carbon electrode (GCE), multi-walled carbon nanotube-graphene oxide has been developed. Covalent organic frameworks (COFs) are dropped with gold nanoparticles (AuNPs), secondary antibody and thionine (Thi-Ab2-Au-COFs) act as the signal probe with high electronic conductivity. Under the ideal conditions, the immunosensor displayed a broad linear response range from 0.01 ng mL-1 to 100 ng mL-1, with a detection limit of 50 pg mL-1 (S/N = 3). The immunosensor also demonstrates outstanding sensitivity, repeatability, and stability. Finally, we utilized the designed immunosensor to detect plasma LXN in coronary artery disease (CAD) patients, and the data showed that plasma LXN was significantly increased in CAD patients with a good performance of ROCAUC (AUC 0.871, 95 % CI 0.725-1.0, p = 0.002), indicating plasma LXN is a potential biomarker of cardiovascular disease. This immunosensor is a promising strategy for screening CAD patients in clinical practice.


Subject(s)
Biosensing Techniques , Cardiovascular Diseases , Coronary Artery Disease , Graphite , Metal Nanoparticles , Metal-Organic Frameworks , Humans , Coronary Artery Disease/diagnosis , Gold , Immunoassay/methods , Biosensing Techniques/methods , Biomarkers , Electrochemical Techniques/methods , Limit of Detection
4.
Int J Biol Sci ; 19(14): 4644-4656, 2023.
Article in English | MEDLINE | ID: mdl-37781042

ABSTRACT

Anthracyclines are a class of conventionally and routinely used first-line chemotherapy drugs for cancer treatment. In addition to the direct cytotoxic effects, increasing evidence indicates that the efficacy of the drugs also depends on immunomodulatory effects with unknown mechanisms. Galectin-9 (Gal-9), a member of the ß-galactoside-binding protein family, has been demonstrated to induce T-cell death and promote immunosuppression in the tumor microenvironment. Here, we asked whether anthracycline-mediated immunomodulatory activity might be related to Gal-9. We found that combining doxorubicin with anti-Gal-9 therapy significantly inhibited tumor growth and prolonged overall survival in immune-competent syngeneic mouse models. Moreover, Gal-9 expression was increased in response to doxorubicin in various human and murine cancer cell lines. Mechanistically, doxorubicin induced tumoral Gal-9 by activating the STING/interferon ß pathway. Clinically, Gal-9 and p-STING levels were elevated in the tumor tissues of breast cancer patients treated with anthracyclines. Our study demonstrates Gal-9 upregulation in response to anthracyclines as a novel mechanism mediating immune escape and suggests targeting Gal-9 in combination with anthracyclines as a promising therapeutic strategy for cancer treatment.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Mice , Animals , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Galectins , Neoplasms/drug therapy , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Tumor Microenvironment
5.
Am J Cancer Res ; 13(4): 1209-1239, 2023.
Article in English | MEDLINE | ID: mdl-37168336

ABSTRACT

Nuclear epidermal growth factor receptor (EGFR) has been shown to be correlated with drug resistance and a poor prognosis in patients with cancer. Previously, we have identified a tripartite nuclear localization signal (NLS) within EGFR. To comprehensively determine the functions and underlying mechanism of nuclear EGFR and its clinical implications, we aimed to explore the nuclear export signal (NES) sequence of EGFR that is responsible for interacting with the exportins. We combined in silico prediction with site-directed mutagenesis approaches and identified a putative NES motif of EGFR, which is located in amino acid residues 736-749. Mutation at leucine 747 (L747) in the EGFR NES led to increased nuclear accumulation of the protein via a less efficient release of the exportin CRM1. Interestingly, L747 with serine (L747S) and with proline (L747P) mutations were found in both tyrosine kinase inhibitor (TKI)-treated and -naïve patients with lung cancer who had acquired or de novo TKI resistance and a poor outcome. Reconstituted expression of the single NES mutant EGFRL747P or EGFRL747S, but not the dual mutant along with the internalization-defective or NLS mutation, in lung cancer cells promoted malignant phenotypes, including cell migration, invasiveness, TKI resistance, and tumor initiation, supporting an oncogenic role of nuclear EGFR. Intriguingly, cells with germline expression of the NES L747 mutant developed into B cell lymphoma. Mechanistically, nuclear EGFR signaling is required for sustaining nuclear activated STAT3, but not for Erk. These findings suggest that EGFR functions are compartmentalized and that nuclear EGFR signaling plays a crucial role in tumor malignant phenotypes, leading to tumorigenesis in human cancer.

6.
Am J Cancer Res ; 12(10): 4721-4736, 2022.
Article in English | MEDLINE | ID: mdl-36381324

ABSTRACT

N-linked glycosylation of proteins is one of the post-translational modifications (PTMs) that shield tumor antigens from immune attack. Signaling lymphocytic activation molecule family 7 (SLAMF7) suppresses cancer cell phagocytosis and is an ideal target under clinical development. PTM of SLAMF7, however, remains less understood. In this study, we investigated the role of N-glycans on SLAMF7 in breast cancer progression. We identified seven N-linked glycosylation motifs on SLAMF7, which are majorly occupied by complex structures. Evolutionally conserved N98 residue is enriched with high mannose and sialylated glycans. Hyperglycosylated SLAMF7 was associated with STT3A expression in breast cancer cells. Inhibition of STT3A by a small molecule inhibitor, N-linked glycosylation inhibitor-1 (NGI-1), reduced glycosylation of SLAMF7, resulting in enhancing antibody affinity and phagocytosis. To provide an on-target effect, we developed an antibody-drug conjugate (ADC) by coupling the anti-SLAMF7 antibody with NGI-1. Deglycosylation of SLAMF7 increases antibody recognition and promotes macrophage engulfment of breast cancer cells. Our work suggests deglycosylation by ADC is a potential strategy to enhance the response of immunotherapeutic agents.

7.
J Am Chem Soc ; 144(23): 10407-10416, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35658433

ABSTRACT

Clinical chemotherapeutic drugs have occasionally been observed to induce antitumor immune responses beyond the direct cytotoxicity. Such effects are coined as immunogenic cell death (ICD), representing a "second hit" from the host immune system to tumor cells. Although chemo-immunotherapy is highly promising, ICD inducers remain sparse with vague drug-target mechanisms. Here, we report an endoplasmic reticulum stress-inducing cyclometalated Ir(III)-bisNHC complex (1a) as a new ICD inducer, and based on this compound, a clickable photoaffinity probe was designed for target identification, which unveiled the engagement of the master regulator protein BiP (binding immunoglobulin protein)/GRP78 of the unfolded protein response pathway. This has been confirmed by a series of cellular and biochemical studies including fluorescence microscopy, cellular thermal shift assay, enzymatic assays, and so forth, showing the capability of 1a for BiP destabilization. Notably, besides 1a, the previously reported ICD inducers including KP1339, mitoxantrone, and oxaliplatin were also found to engage BiP interaction, suggesting the important role of BiP in eliciting anticancer immunity. We believe that the ICD-related target information in this work will help to understand the mode of action of ICD that is beneficial to designing new ICD agents with high specificity and improved efficacy.


Subject(s)
Antineoplastic Agents , Immunogenic Cell Death , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Iridium/pharmacology , Unfolded Protein Response
8.
EBioMedicine ; 74: 103712, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34839261

ABSTRACT

BACKGROUND: Despite clinical success with anti-spike vaccines, the effectiveness of neutralizing antibodies and vaccines has been compromised by rapidly spreading SARS-CoV-2 variants. Viruses can hijack the glycosylation machinery of host cells to shield themselves from the host's immune response and attenuate antibody efficiency. However, it remains unclear if targeting glycosylation on viral spike protein can impair infectivity of SARS-CoV-2 and its variants. METHODS: We adopted flow cytometry, ELISA, and BioLayer interferometry approaches to assess binding of glycosylated or deglycosylated spike with ACE2. Viral entry was determined by luciferase, immunoblotting, and immunofluorescence assays. Genome-wide association study (GWAS) revealed a significant relationship between STT3A and COVID-19 severity. NF-κB/STT3A-regulated N-glycosylation was investigated by gene knockdown, chromatin immunoprecipitation, and promoter assay. We developed an antibody-drug conjugate (ADC) that couples non-neutralization anti-spike antibody with NGI-1 (4G10-ADC) to specifically target SARS-CoV-2-infected cells. FINDINGS: The receptor binding domain and three distinct SARS-CoV-2 surface N-glycosylation sites among 57,311 spike proteins retrieved from the NCBI-Virus-database are highly evolutionarily conserved (99.67%) and are involved in ACE2 interaction. STT3A is a key glycosyltransferase catalyzing spike glycosylation and is positively correlated with COVID-19 severity. We found that inhibiting STT3A using N-linked glycosylation inhibitor-1 (NGI-1) impaired SARS-CoV-2 infectivity and that of its variants [Alpha (B.1.1.7) and Beta (B.1.351)]. Most importantly, 4G10-ADC enters SARS-CoV-2-infected cells and NGI-1 is subsequently released to deglycosylate spike protein, thereby reinforcing the neutralizing abilities of antibodies, vaccines, or convalescent sera and reducing SARS-CoV-2 variant infectivity. INTERPRETATION: Our results indicate that targeting evolutionarily-conserved STT3A-mediated glycosylation via an ADC can exert profound impacts on SARS-CoV-2 variant infectivity. Thus, we have identified a novel deglycosylation method suitable for eradicating SARS-CoV-2 variant infection in vitro. FUNDING: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.


Subject(s)
Benzamides/pharmacology , COVID-19 Drug Treatment , Glycosylation/drug effects , Hexosyltransferases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Virus Internalization/drug effects , A549 Cells , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , HEK293 Cells , Hexosyltransferases/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , SARS-CoV-2/growth & development , Spike Glycoprotein, Coronavirus/metabolism
10.
J Inorg Biochem ; 213: 111260, 2020 12.
Article in English | MEDLINE | ID: mdl-33039746

ABSTRACT

Two copper complexes, Cu1 (CuL1Cl2, L1 = 2-(6,7-dimethoxyisoquinolin-1-yl) aniline) and Cu2 (CuL2Cl2, L2 = 2-(6-methoxyisoquinolin-1-yl) aniline), were synthesized and characterized. These complexes exhibited high cytotoxic activity toward different cancer cell lines, including the A549 lung cancer cell line, and low cytotoxicity toward normal human cells. Mechanistic studies have shown that these complexes induce bimodal death of cancer cells through apoptosis and autophagy, including the activation of apoptotic and autophagic cell signaling pathways. In addition, Cu1 and Cu2 interacted with calf thymus DNA (ct-DNA) via an intercalative binding mode. The different biological behaviors of these copper complexes could be attributed to the presence of electron-donating methoxy groups on the ligands. Cu1 and Cu2 effectively inhibited tumor growth in a xenografted mouse model bearing A549 cells but exhibited lower in vivo toxicity than cisplatin. Thus, Cu1 and Cu2 can be developed as potential anticancer agents.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Coordination Complexes/pharmacology , Copper/pharmacology , A549 Cells , Animals , Binding, Competitive , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System/drug effects , Mice , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
11.
Nat Cell Biol ; 22(10): 1264-1275, 2020 10.
Article in English | MEDLINE | ID: mdl-32929201

ABSTRACT

Although pyroptosis is critical for macrophages against pathogen infection, its role and mechanism in cancer cells remains unclear. PD-L1 has been detected in the nucleus, with unknown function. Here we show that PD-L1 switches TNFα-induced apoptosis to pyroptosis in cancer cells, resulting in tumour necrosis. Under hypoxia, p-Stat3 physically interacts with PD-L1 and facilitates its nuclear translocation, enhancing the transcription of the gasdermin C (GSDMC) gene. GSDMC is specifically cleaved by caspase-8 with TNFα treatment, generating a GSDMC N-terminal domain that forms pores on the cell membrane and induces pyroptosis. Nuclear PD-L1, caspase-8 and GSDMC are required for macrophage-derived TNFα-induced tumour necrosis in vivo. Moreover, high expression of GSDMC correlates with poor survival. Antibiotic chemotherapy drugs induce pyroptosis in breast cancer. These findings identify a non-immune checkpoint function of PD-L1 and provide an unexpected concept that GSDMC/caspase-8 mediates a non-canonical pyroptosis pathway in cancer cells, causing tumour necrosis.


Subject(s)
Apoptosis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Pyroptosis , Animals , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Female , Humans , Hypoxia/physiopathology , Inflammasomes , Mice , Mice, Inbred BALB C , Mice, Nude , Necrosis , Neoplasms/genetics , Neoplasms/metabolism , Tumor Cells, Cultured , Tumor-Associated Macrophages , Xenograft Model Antitumor Assays
12.
Biol Trace Elem Res ; 195(2): 472-480, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31444770

ABSTRACT

Chronic copper exposure impaired spermatogenesis in adult male mice. The aim of this study was to determine whether chronic copper exposure can induce apoptosis of testicular cell and hypospermatogenesis via disturbing testosterone synthesis in adult male mice. In the present study, sixty CD-1 male mice were randomly divided into four groups, and were continuously administered for 8 weeks by oral gavage with copper sulfate at a dose of 0, 25, 100, and 150 mg/kg/day, respectively. We determined the content of serum and testicular copper, testicular coefficient, testicular histopathology, sperm count and motility, the mRNA and protein levels of Caspase-3, Bax, and Bcl-2, Leydig cell count, testosterone content, testosterone synthetase, and testosterone synthesis-related genes. The results showed that the copper levels in serum increased in a dose-dependent manner, and the copper levels in testes were significantly related to serum copper levels. Male mice given copper sulfate 100 and 150 dosage groups showed significant decreased in sperm motility and sperm number as well as increased in testes damage, and there was no significant change in testicular coefficient in the four groups. The mRNA levels of Bcl-2 decreased and Caspase-3 increased in 150 dosage group, and Bax increased in two higher dosage groups. Meanwhile, Caspase-3 and Bax proteins increased in 150 dosage group, and Bcl-2 protein decreased in three copper treatment groups. Nevertheless, there were no differences on the levels of testosterone content and testosterone synthetase of 3ß-HSD, 17ß-HSD, 17α-Hyd, and 20α-Hyd, mRNA levels of Cyp11a1, Cyp17a1, and Star, and quantity of Leydig cells in four groups. Overall, these data showed that chronic copper exposure led to copper residues in the testes, and the doses of 100 and 150 mg/kg/day copper sulfate may induce hypospermatogenesis by increasing apoptosis without affecting testosterone secretion.


Subject(s)
Apoptosis/drug effects , Copper Sulfate/pharmacology , Spermatogenesis/drug effects , Testis/drug effects , Testosterone/metabolism , Administration, Oral , Animals , Copper Sulfate/administration & dosage , Copper Sulfate/analysis , Male , Mice , Testis/metabolism , Testis/pathology , Testosterone/blood
13.
Chem Commun (Camb) ; 55(87): 13066-13069, 2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31570904

ABSTRACT

A platinum(ii) complex containing an aminophosphonate ligand preferentially accumulates in the endoplamic reticulum (ER) in association with potent ER stress and reactive oxygen species generation, followed by the activation of damage-associated molecular pattern signals and immune responses. Importantly, the Pt complex exhibits potent anti-tumour activities in two independent mouse models via an immunogenic cell death pathway.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Esters/pharmacology , Lung Neoplasms/drug therapy , Organophosphonates/pharmacology , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents, Immunological/chemistry , Cell Death/drug effects , Esters/chemistry , Humans , Ligands , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Organophosphonates/chemistry , Organoplatinum Compounds/chemistry
14.
Org Lett ; 21(13): 4929-4932, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31082239

ABSTRACT

An efficient strategy for the synthesis of benzofuro[2,3- b]pyrazines was developed. These tricyclic scaffolds were formed through a multistep cascade sequence, which includes double insertion of isonitriles and chemoselective bicyclization. In this reaction, a nanopalladium was used as a recyclable catalyst. Product 3w exhibited excellent anticancer activity toward T-24 (IC50 = 12.5 ± 0.9 µM) and HeLa (IC50 = 14.7 ± 1.6 µM) cells. We also explored the action mechanism of 3w on T-24 cells.


Subject(s)
Nitriles/chemistry , Nitriles/chemical synthesis , Phenols/chemistry , Phenols/chemical synthesis , Pyrazines/chemistry , Pyrazines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Catalysis , Chemistry Techniques, Synthetic , Porosity
15.
Gene ; 688: 19-25, 2019 Mar 10.
Article in English | MEDLINE | ID: mdl-30447345

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer in adults and is a leading cause of worldwide cancer mortality. Intrahepatic dissemination and extrahepatic metastasis are key factors in malignant growth of HCC. Reducing HCC-associated metastasis is critically dependent on uncovering molecular signaling pathways that promote HCC metastasis. In this study, we explored the effect of TGF-ß1 and RELN on cell migration, and the relationship between TGF-ß1 and RELN in HCC cells. The data presented that TGF-ß1 and RELN showed an opposite expression pattern, and either increased expression of TGF-ß1 or decreased expression of RELN increased HCC cell migration ability. We also found TGF-ß1 enhanced cell migration ability was through repressing RELN expression, as overexpression of RELN impaired TGF-ß1 enhanced cell migration. Our work revealed the relationship between TGF-ß1 and RELN and uncovered the important role of RELN in suppressing cell migration in HCC cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Hep G2 Cells , Humans , Reelin Protein , Signal Transduction/physiology
16.
Free Radic Biol Med ; 129: 418-429, 2018 12.
Article in English | MEDLINE | ID: mdl-30266678

ABSTRACT

Agents with multiple modes of tumor cell death can be effective chemotherapeutic drugs. One example of a bimodal chemotherapeutic approach is an agent that can induce both apoptosis and autophagic death. Thus far, no clinical anticancer drug has been shown to simultaneously induce both these pathways. Mono-functional platinum complexes are potent anticancer drug candidates which act through mechanisms distinct from cisplatin. Here, we describe the synthesis and characterize of two mono-functional platinum complexes containing 8-substituted quinoline derivatives as ligands. In comparison to cisplatin, n-Mon-Pt-1 exhibited a greater in vitro cytotoxicity, was more effective in resistant cells and elicited a better anticancer effect. Mechanistic experiments indicate that n-Mon-Pt-1 mainly accumulates in mitochondria, and stimulates significant TrxR inhibition, ROS release and an ER stress response, ultimately resulting in a simultaneous induction of apoptosis and autophagy. Importantly, compared to cisplatin, n-Mon-Pt-1 exhibits lower acute toxicity and better anticancer activity in a murine tumor model.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , A549 Cells , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/genetics , Autophagy/genetics , Cell Line , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Organoplatinum Compounds/chemical synthesis , Quinolines/chemistry , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
17.
Eur J Med Chem ; 155: 639-650, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29935437

ABSTRACT

Agents with multiple modes of tumor cell death can be effective chemotherapeutic drugs. One example of a bimodal chemotherapeutic approach is an agent that can induce both apoptosis and autophagic death. Thus far, no clinical anticancer drug has been shown to simultaneously induce both these pathways. Mono-functional platinum complexes are potent anticancer drug candidates which act through mechanisms distinct from cisplatin. Here, we describe the synthesis and characterize of two mono-functional platinum complexes containing 8-substituted quinoline derivatives as ligands, [PtL1Cl]Cl [L1 = (Z)-1-(pyridin-2-yl)-N-(quinolin-8-ylmethylene) methanamine] (Mon-Pt-1) and [PtL2Cl]Cl [L2 = (Z)-2-(pyridin-2-yl)-N-(quinolin-8-ylmethylene) ethanamine] (Mon-Pt-2). In comparison to cisplatin, Mon-Pt-2 exhibited a greater in vitro cytotoxicity, was more effective in resistant cells and elicited a better anticancer effect. Mechanistic experiments indicate that Mon-Pt-2 mainly accumulates in mitochondria, and stimulates significant TrxR inhibition ROS release and an ER stress response, mediated by mitochondrial dysfunction, ultimately resulting in a simultaneous induction of apoptosis and autophagy. Importantly, compared to cisplatin, Mon-Pt-2 exhibits lower acute toxicity and better anticancer activity in a murine tumor model. To the best of our knowledge, Mon-Pt-2 is the first mono-functional platinum complex inducing pro-death autophagy and apoptosis of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Mitochondria/drug effects , A549 Cells , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Humans , Mice , Mice, Inbred Strains , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Structure-Activity Relationship
18.
J Med Chem ; 61(8): 3478-3490, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29606001

ABSTRACT

Agents inducing both apoptosis and autophagic death can be effective chemotherapeutic drugs. In our present work, we synthesized two organometallic gold(III) complexes harboring C^N ligands that structurally resemble tetrahydroisoquinoline (THIQ): Cyc-Au-1 (AuL1Cl2, L1 = 3,4-dimethoxyphenethylamine) and Cyc-Au-2 (AuL2Cl2, L2 = methylenedioxyphenethylamine). In screening their in vitro activity, we found both gold complexes exhibited lower toxicity, lower resistance factors, and better anticancer activity than those of cisplatin. The organometallic gold(III) complexes accumulate in mitochondria and induce elevated ROS and an ER stress response through mitochondrial dysfunction. These effects ultimately result in simultaneous apoptosis and autophagy. Importantly, compared to cisplatin, Cyc-Au-2 exhibits lower toxicity and better anticancer activity in a murine tumor model. To the best of our knowledge, Cyc-Au-2 is the first organometallic Au(III) compound that induces apoptosis and autophagic death. On the basis of our results, we believe Cyc-Au-2 to be a promising anticancer agent or lead compound for further anticancer drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Coordination Complexes/pharmacology , Endoplasmic Reticulum Stress/drug effects , Tetrahydroisoquinolines/pharmacology , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Coordination Complexes/chemical synthesis , Coordination Complexes/therapeutic use , Coordination Complexes/toxicity , Drug Screening Assays, Antitumor , Humans , Ligands , Male , Mice, Nude , Mitochondria/drug effects , Molecular Structure , Reactive Oxygen Species/metabolism , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/therapeutic use , Tetrahydroisoquinolines/toxicity , Xenograft Model Antitumor Assays
19.
Oncotarget ; 8(35): 59359-59375, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938642

ABSTRACT

Lysicamine is a natural oxoaporphine alkaloid, which isolated from traditional Chinese medicine (TCM) herbs and has been shown to possess cytotoxicity to hepatocarcinoma cell lines. Reports on its antitumor activity are scarce because lysicamine occurs in plants at a low content. In this work, we demonstrate a facile concise total synthesis of lysicamine from simple raw materials under mild reaction conditions, and the preparation of the Ru(II), Rh(III), Mn(II) and Zn(II) complexes 1-4 of lysicamine (LY). All the compounds were fully characterized by elemental analysis, IR, ESI-MS, 1H and 13C NMR, as well as single-crystal X-ray diffraction analysis. Compared with the free ligand LY, complexes 2 and 3 exhibited superior in vitro cytotoxicity against HepG2 and NCI-H460. Mechanistic studies indicated that 2 and 3 blocked the cell cycle in the S phase by decreasing of cyclins A2/B1/D1/E1, CDK 2/6, and PCNA levels and increasing levels of p21, p27, p53 and CDC25A proteins. In addition, 2 and 3 induced cell apoptosis via both the caspase-dependent mitochondrial pathway and the death receptor pathway. in vivo study showed that 2 inhibited HepG2 tumor growth at 1/3 maximum tolerated dose (MTD) and had a better safety profile than cisplatin.

20.
J Inorg Biochem ; 169: 23-31, 2017 04.
Article in English | MEDLINE | ID: mdl-28095346

ABSTRACT

Four µ2-Cl bridged dinuclear metal complexes with isoquinoline ligands, (MPDQ)2Zn2Cl4 (1) (MPDQ=4.5-methylenedioxy-1-pyridinedihydroisoquinoline), (PYP)2Zn2Cl4 (2) (PYP=5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline), (MPDQ)2Mn2Cl4 (3),and (PYP)2Mn2Cl4 (4) were synthesized and characterized. All complexes exhibited strong proliferation inhibition activity against various cancer cells. The underlying molecular mechanisms through which they caused the cancer cell death were also elucidated. Induction of apoptosis in MGC-803 cells by complex 2 was evidenced by annexin V+/PI- detection and DiD/DAPI staining assay. Further investigation revealed that complex 2 was able to induce intrinsic pathway-dependent apoptosis in cancer cells by triggering DNA damage which was caused by the overproduction of reactive oxygen species. Based on these studies, we suggest that Zn(II) complexes containing isoquinoline ligands can be developed as candidates for anti-cancer chemotherapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Isoquinolines/chemistry , Manganese/chemistry , Zinc/chemistry , A549 Cells , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...