Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 268: 116301, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38452727

ABSTRACT

In this work, a novel of dual tubulin/HDAC inhibitors were designed and synthesized based on the structure of natural product millepachine, which has been identified as a tubulin polymerization inhibitor. Biological evaluation revealed that compound 9n exhibited an impressive potency against PC-3 cells with the IC50 value of 16 nM and effectively inhibited both microtubule polymerization and HDAC activity. Furthermore, compound 9n not only induced cell cycle arrest at G2/M phase, but also induced PC- 3 cells apoptosis. Further study revealed that the induction of cell apoptosis by 9n was accompanied by a decrease in mitochondrial membrane potential and an elevation in reactive oxygen species levels in PC-3 cells. Additionally, 9n exhibited inhibitory effects on tumor cell migration and angiogenesis. In PC-3 xenograft model, 9n achieved a remarkable tumor inhibition rate of 90.07%@20 mg/kg, significantly surpassing to that of CA-4 (55.62%@20 mg/kg). Meanwhile, 9n exhibited the favorable drug metabolism characteristics in vivo. All the results indicate that 9n is a promising dual tubulin/HDAC inhibitor for chemotherapy of prostate cancer, deserving the further investigation.


Subject(s)
Antineoplastic Agents , Chalcones , Prostatic Neoplasms , Male , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Tubulin Modulators/chemistry , Histone Deacetylase Inhibitors/pharmacology , Cell Line, Tumor , Structure-Activity Relationship , Tubulin/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Cell Proliferation , Prostatic Neoplasms/drug therapy , Apoptosis
2.
J Control Release ; 368: 780-796, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499091

ABSTRACT

Designing effective nanomedicines to induce durable anti-tumor immunity represents a promising strategy for improving moderate immune stimulation. In this study, we engineered a multifunctional nanoreactor (named SCGFP NPs) for remodeling the tumor microenvironment (TME) to improve the therapeutic efficacy of immunotherapy. The core of SCGFP NPs consists of CaCO3 loaded with SN38, prepared by the gas diffusion method, and coated with a significant amount of gallic acid-Fe3+-PEG coordination polymer on the surface. In the acidic TME, SCGFP NPs explosively release exogenous Ca2+ and SN38. The SN38-induced intracellular Ca2+ accumulation and exogenous Ca2+ synergistically trigger immunogenic cell death (ICD) through sustained Ca2+ overload. The ablation of tumors with high-intensity photothermal therapy (PTT) by near-infrared (NIR) irradiation of GA-Fe3+ induces tumor cell necrosis, further enhancing ICD activation. Additionally, SN38 upregulates PD-L1, amplifying tumor responsiveness to immune checkpoint inhibitors (ICIs). This study indicates that SCGFP NPs, through the integration of a trimodal therapeutic strategy, hold enormous potential for various types of tumor immunotherapy through distinct mechanisms or synergistic effects.


Subject(s)
Immunotherapy , Neoplasms , Bioreactors , Diffusion , Gallic Acid/therapeutic use , Polymers , Tumor Microenvironment , Cell Line, Tumor
3.
J Med Chem ; 67(4): 3144-3166, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38336655

ABSTRACT

Cancer immunotherapy has revolutionized clinical advances in a variety of cancers. Due to the low immunogenicity of the tumor, only a few patients can benefit from it. Specific microtubule inhibitors can effectively induce immunogenic cell death and improve immunogenicity of the tumor. A series of isoquinoline derivatives based on the natural products podophyllotoxin and diphyllin were designed and synthesized. Among them, F10 showed robust antiproliferation activity against four human cancer cell lines, and it was verified that F10 exerted antiproliferative activity by inhibiting tubulin and V-ATPase. Further studies indicated that F10 is able to induce immunogenic cell death in addition to apoptosis. Meanwhile, F10 inhibited tumor growth in an RM-1 homograft model with enhanced T lymphocyte infiltration. These results suggest that F10 may be a promising lead compound for the development of a new generation of microtubule drugs.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Tubulin/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Structure-Activity Relationship , Polymerization , Adenosine Triphosphatases/metabolism , Immunogenic Cell Death , Drug Screening Assays, Antitumor , Neoplasms/drug therapy , Neoplasms/metabolism , Apoptosis , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Cell Proliferation , Cell Line, Tumor
4.
Eur J Med Chem ; 268: 116204, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38364716

ABSTRACT

The involvement of CDC20 in promoting tumor growth in different types of human cancers and it disturbs the process of cell division and impedes tumor proliferation. In this work, a novel of Apcin derivatives targeting CDC20 were designed and synthesized to evaluate for their biological activities. The inhibitory effect on the proliferation of four human tumor cell lines (MCF-7, MDA-MB-231, MDA-MB-468 and A549) was observed. Among them, compound E1 exhibited the strongest inhibitory effect on the proliferation of MDA-MB-231 cells with an IC50 value of 1.43 µM, which was significantly superior to that of Apcin. Further biological studies demonstrated that compound E1 inhibited cancer cell migration and colony formation. Furthermore, compound E1 specifically targeted CDC20 and exhibited a higher binding affinity to CDC20 compared to that of Apcin, thereby inducing cell cycle arrest in the G2/M phase of cancer cells. Moreover, it has been observed that compound E1 induces autophagy in cancer cells. In 4T1 Xenograft Models compound E1 exhibited the potential antitumor activity without obvious toxicity. These findings suggest that E1 could be regarded as a CDC20 inhibitor deserved further investigation.


Subject(s)
Antineoplastic Agents , Diamines , Triple Negative Breast Neoplasms , Humans , Cell Proliferation , Triple Negative Breast Neoplasms/pathology , Apoptosis , Carbamates/pharmacology , Cell Line, Tumor , Cell Cycle Proteins , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Cdc20 Proteins
5.
J Med Chem ; 66(14): 10036-10059, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37452764

ABSTRACT

Glutathione peroxidase 4 (GPX4) is a promising target to induce ferroptosis for the treatment of triple-negative breast cancer (TNBC). We designed and synthesized a novel series of covalent GPX4 inhibitors based on RSL3 and ML162 by structural integration and simplification strategies. Among them, compound C18 revealed a remarkable inhibitory activity against TNBC cells and significantly inhibited the activity of GPX4 compared to RSL3 and ML162. Moreover, it was identified that C18 could notably induce ferroptosis with high selectivity by increasing the accumulation of lipid peroxides (LPOs) in cells. Further study demonstrated that C18 covalently bound to the Sec46 of GPX4. Surprisingly, C18 exhibited an outstanding potency of tumor growth inhibition in the MDA-MB-231 xenograft model with a TGI value of 81.0%@20 mg/kg without obvious toxicity. Overall, C18 could be a promising GPX4 covalent inhibitor to induce ferroptosis for the treatment of TNBC.


Subject(s)
Ferroptosis , Triple Negative Breast Neoplasms , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , Glutathione Peroxidase/metabolism , Triple Negative Breast Neoplasms/drug therapy , Lipid Peroxides
6.
J Med Chem ; 65(24): 16774-16800, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36471625

ABSTRACT

Natural products are a major source of anticancer agents and play critical roles in anticancer drug development. Inspired by the complexity-to-diversity strategy, novel deoxypodophyllotoxin (DPT) analogues were designed and synthesized. Among them, compound C3 exhibited the potent antiproliferative activity against four human cancer cell lines with IC50 values in the low nanomolar range. Additionally, it showed marked activity against paclitaxel-resistant MCF-7 cells and A549 cells. Moreover, compound C3 can inhibit tubulin polymerization by targeting the colchicine-binding site of tubulin. Further study revealed that compound C3 could arrest cancer cells in the G2/M phase and disrupt the angiogenesis in human umbilical vein endothelial cells. Meanwhile, C3 remarkably inhibited cancer cell motility and migration, as well as considerably inhibited tumor growth in MCF-7 and MCF-7/TxR xenograft model without obvious toxicity. Collectively, these results indicated that compound C3 may be a promising tubulin polymerization inhibitor development for cancer treatment.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/therapeutic use , Tubulin Modulators/chemistry , Colchicine/metabolism , Tubulin/metabolism , Endothelial Cells/metabolism , Drug Screening Assays, Antitumor , Cell Proliferation , Binding Sites , MCF-7 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Polymerization , Cell Line, Tumor , Structure-Activity Relationship , Neoplasms/drug therapy
7.
Eur J Med Chem ; 237: 114372, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35447432

ABSTRACT

A novel series of diphenylamine derivatives were designed and synthesized, and their biological activities were evaluated. The anti-proliferative activities of the derivatives were tested against five human cancer cell lines (MCF-7, MDA-MB-231, A549, HeLa and HT29). Among them, compound 5f exhibited the promising anti-proliferative activity against HT29 cell lines with the IC50 value of 23 nM. Further biological studies depicted that compound 5f inhibited cancer cell migration, colony formation and angiogenesis. Besides, dynamics studies and molecular docking studies revealed that compound 5f inhibited tubulin polymerization which may be a result of the compound binding to the colchicine site of tubulin. Furthermore, compound 5f arrested HT29 cell cycle at G2/M phase, and induced HT29 cell apoptosis by upregulating cyclin B1, Bcl-2, Bax, Cleaved-caspase9, Cleaved-caspase3, PARP, Cleaved-PARP proteins, and downregulating p-cdc25c (S216), p-cdc2 (T15) proteins. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) were also determined to confirm the cell apoptosis process. Finally, compound 5f greatly inhibited the tumor growth in HT29 xenograft mice by 75.5% at 10 mg/kg. Meanwhile, compound 5f owned the good pharmacokinetic properties. All the results promised that 5f is of potential to act as an antitumor candidate and worthy of further investigation.


Subject(s)
Antineoplastic Agents , Tubulin Modulators , Animals , Antineoplastic Agents/chemistry , Binding Sites , Cell Line, Tumor , Cell Proliferation , Colchicine/pharmacology , Diphenylamine/pharmacology , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Docking Simulation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Polymerization , Tubulin/metabolism , Tubulin Modulators/chemistry
8.
Eur J Med Chem ; 190: 112105, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32035399

ABSTRACT

A novel series of shikonin-benzo[b]furan derivatives were designed and synthesized as tubulin polymerization inhibitors, and their biological activities were evaluated. Most compounds revealed the comparable anti-proliferation activities against the cancer cell lines to that of shikonin and simultaneously low cytotoxicity to non-cancer cells. Among them, compound 6c displayed powerful anti-cancer activity with the IC50 value of 0.18 µM against HT29 cells, which was significantly better than that of the reference drugs shikonin and CA-4. What's more, 6c could inhibit tubulin polymerization and compete with [3H] colchicine in binding to tubulin. Further biological studies depicted that 6c can induce cell apoptosis and cell mitochondria depolarize, regulate the expression of apoptosis related proteins in HT29 cells. Besides, 6c actuated the HT29 cell cycle arrest at G2/M phase, and influenced the expression of the cell-cycle related protein. Moreover, 6c displayed potent inhibition on cell migration and tube formation that contributes to the antiangiogenesis. These results prompt us to consider 6c as a potential tubulin polymerization inhibitor and is worthy for further study.


Subject(s)
Benzofurans/pharmacology , Naphthoquinones/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Benzofurans/chemical synthesis , Benzofurans/metabolism , Benzofurans/toxicity , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Molecular Docking Simulation , Naphthoquinones/chemical synthesis , Naphthoquinones/metabolism , Naphthoquinones/toxicity , Protein Binding , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism , Tubulin Modulators/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...