1
|
Fan HL, Chen JL, Liu ST, Lee JT, Huang SM, Wu ZF, Lai HC. Remimazolam induced cytotoxicity mediated through multiple stress pathways and acted synergistically with tyrosine kinase inhibitors in hepatocellular carcinoma. Redox Rep 2025; 30:2475696. [PMID: 40053437 DOI: 10.1080/13510002.2025.2475696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
The primary treatment for hepatocellular carcinoma (HCC) involves surgical removal of the primary tumor, but this creates a favorable environment for the proliferation and spread of residual and circulating cancer cells. The development of remimazolam-based balanced anesthesia is crucial for future antitumor applications. It is important to understand the mechanisms of cytotoxicity for HCC in detail. We performed cell viability analysis, western blotting analysis, reverse transcription-polymerase chain reaction analysis, and flow cytometry analysis in two HCC cell lines, HepG2 and Hep3B cells. Our data demonstrated that remimazolam induced cytotoxicity by suppressing cell proliferation, inhibiting G1 phase progression, and affecting mitochondrial reactive oxygen species (ROS) levels, leading to apoptosis, DNA damage, cytosolic ROS elevation, lipid peroxidation, autophagy, mitochondrial depolarization, and endoplasmic reticulum stress. Inhibitors of apoptosis, autophagic cell death, and ferroptosis and a ROS scavenger failed to rescue cell death caused by remimazolam besylate. Our combination index revealed that remimazolam besylate has the potential to act as a sensitizer for targeted tyrosine kinase inhibitor therapy for HCC. Our findings open up new possibilities for combinatory HCC therapy using remimazolam, leveraging its dual functional roles in surgery and drug therapy for liver cancers.
Collapse
Affiliation(s)
- Hsiu-Lung Fan
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Jia-Lin Chen
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Shu-Ting Liu
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Jia-Tong Lee
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, Republic of China
| | - Zhi-Fu Wu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan, Republic of China
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan, Republic of China
- Center for Regional Anesthesia and Pain Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan, Republic of China
| | - Hou-Chuan Lai
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
2
|
Elkattan HH, Elsisi AE, El-Lakkany NM. Gossypol enhances ponatinib's cytotoxicity against human hepatocellular carcinoma cells by involving cell cycle arrest, p-AKT/LC3II/p62, and Bcl2/caspase-3 pathways. Toxicol Rep 2025; 14:101856. [PMID: 39802605 PMCID: PMC11719416 DOI: 10.1016/j.toxrep.2024.101856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/30/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025] Open
Abstract
Despite significant breakthroughs in frontline cancer research and chemotherapy for hepatocellular carcinoma (HCC), many of the suggested drugs have high toxic side effects and resistance, limiting their clinical utility. Exploring potential therapeutic targets or novel combinations with fewer side effects is therefore crucial in combating this dreadful disease. The current study aims to use a novel combination of ponatinib and gossypol against the HepG2 cell line. Cell survival, FGF19/FGFR4, apoptotic and autophagic cell death, and synergistic drug interactions were assessed in response to increasing concentrations of ponatinib and/or gossypol treatment. Research revealed that ponatinib (1.25-40 μM) and gossypol (2.5-80 μM) reduced the viability of HepG2 cells in a way that was dependent on both time and dose. Ponatinib's anti-proliferation effectiveness was improved synergistically by gossypol and was associated with a rise in apoptotic cell death, cell cycle blockage during the G0/G1 phase, and suppression of the FGF19/FGFR4 axis. Furthermore, the ponatinib/gossypol combination lowered Bcl-2 and p-Akt while increasing active caspase-3, Beclin-1, p62, and LC3II. This combination, however, had no harm on normal hepatocytes. Overall, gossypol enhanced ponatinib's anticancer effects in HCC cells. Notably, this new combination appears to be potential adjuvant targeted chemotherapy, a discovery that warrants more clinical investigation, in the management of patients with HCC.
Collapse
Affiliation(s)
- Hadeel H. Elkattan
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, Giza 12411, Egypt
| | - Alaa E. Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Naglaa M. El-Lakkany
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, Giza 12411, Egypt
| |
Collapse
|
3
|
Zhang J, Li S, Hou H, Lu X. A novel mathematical model for studying antimicrobial interactions against viable but non-culturable Campylobacter jejuni in the poultry product processing environment. Food Microbiol 2025; 128:104740. [PMID: 39952754 DOI: 10.1016/j.fm.2025.104740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/06/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Campylobacter jejuni is a major pathogen associated with gastrointestinal illness and is frequently detected in poultry products. Its ability to enter the viable but non-culturable (VBNC) state is an adaptive survival strategy triggered by adverse conditions. Consequently, the processing conditions involved in poultry production can potentially induce C. jejuni into the VBNC state, posing risks to food safety and public health. This study aimed to evaluate the antimicrobial effectiveness of carvacrol, diallyl sulfide, and Al2O3 nanoparticles (NPs) and investigate their synergistic interactions against VBNC C. jejuni under simulated poultry processing conditions. The time-kill assay demonstrated that Al₂O₃ NPs achieved >1 log CFU/mL reductions at 0.3 mg/mL, while carvacrol and diallyl sulfide required higher concentrations (0.8 mg/mL and 1.6 mg/mL, respectively) to achieve comparable reductions. While additive effects were observed for all combinations, the interactions were further examined using the combination index. The mathematical model effectively simulated the antimicrobial effects and interactions across varying levels of inhibition, confirming the potent antimicrobial activity of Al2O3 NPs. While carvacrol and diallyl sulfide exhibited additive effects in combination, synergistic interactions (combination index <1) were identified for binary and ternary combinations with Al₂O₃ NPs, including carvacrol/Al₂O₃ NPs, diallyl sulfide/Al₂O₃ NPs, and carvacrol/diallyl sulfide/Al₂O₃ NPs. These findings underscore the potential of Al₂O₃ NPs, individually or in combination with plant-based antimicrobials, to mitigate VBNC C. jejuni and improve food safety. The mathematical model presents an alternative approach to developing novel antimicrobial strategies.
Collapse
Affiliation(s)
- Jingbin Zhang
- Beijing Life Science Academy, Beijing, 102209, China; Department of Food Science and Agricultural Chemistry, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Quebec, H9X 3V9, Canada
| | - Shenmiao Li
- Department of Food Science and Agricultural Chemistry, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Quebec, H9X 3V9, Canada
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing, 102209, China
| | - Xiaonan Lu
- Department of Food Science and Agricultural Chemistry, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Quebec, H9X 3V9, Canada.
| |
Collapse
|
4
|
Zhu M, Xie Y, Li Z, Bao H, Miao D, Guo X, Wang S, Chen K, Chen H, Dai J, Yang N, Yu L, Pei J. Antitumour and anti-angiogenesis efficacy of a multifunctional self-oxygenated active-targeting drug delivery system by encapsulating biological and chemotherapeutic drugs. Colloids Surf B Biointerfaces 2025; 250:114549. [PMID: 39965481 DOI: 10.1016/j.colsurfb.2025.114549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
The hypoxic tumour microenvironment (TME), resulting from abnormal tumour angiogenesis, is a major factor contributing to treatment failure in breast cancer patients. In this study, we present a ZnO2-based oestrone-conjugated PEGylated liposome (ZnO2@EPL-CDDP/EGCG) that incorporates cisplatin (CDDP) and epigallocatechin-3-gallate (EGCG). ZnO2 remains stable in neutral environments but decomposes under mildly acidic conditions, releasing Zn²⁺ and H₂O₂. These byproducts inhibit the electron transport chain, stimulate the endogenous reactive oxygen species production for chemodynamic therapy (CDT), and generate oxygen at tumour sites to alleviate hypoxia and enhance anti-angiogenic efficacy. EGCG inhibits tumour angiogenesis by down-regulating hypoxia-inducible factor-1α (HIF-1α) and its downstream pathways, while also exhibiting synergistic anti-tumour effects with CDDP. Oestrone-conjugated and polyethylene glycol (PEG) modifications facilitate targeted accumulation at tumour sites. Our findings indicate that ZnO2@EPL-CDDP/EGCG significantly improves the therapeutic efficacy of both EGCG and CDDP, remodels tumour vasculature, and alleviates hypoxia within the TME. This self-oxygenated, actively targeted drug delivery system notably extends the survival of healthy ICR mice without observed toxicity. This novel approach, which co-encapsulates ZnO2, EGCG, and CDDP in an active-targeting liposomal formulation for the first time, represents a promising strategy for effective cancer treatment.
Collapse
Affiliation(s)
- Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Zhiping Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Kejia Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Hongzhu Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Jingwen Dai
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Liangping Yu
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, PR China.
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China.
| |
Collapse
|
5
|
Hajir S, Jobst KJ, Kleywegt S, Simpson AJ, Simpson MJ. Do co-solvents used in exposure studies equally perturb the metabolic profile of Daphnia magna? ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2025:vgaf068. [PMID: 40246286 DOI: 10.1093/etojnl/vgaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/19/2025]
Abstract
Dissolution methods such as co-solvents are used to solubilize insoluble compounds in exposure experiments. Several exposure studies have followed the guidelines from the Organization for Economic Co-operation and Development where co-solvents are applied at 0.01% v/v of the total exposure volume. Although no observable apical endpoint abnormalities were reported following these guidelines, little is known about the molecular-level impacts of co-solvents used in exposure studies. A targeted metabolomics approach using liquid chromatography coupled with triple quadrupole mass spectrometry was used to assess Daphnia magna responses to four commonly used co-solvents, including acetone (ACT), acetonitrile (ACN), methanol (MeOH), and dimethyl sulfoxide (DMSO), at three different levels (0.01%, 0.05%, and 0.1% v/v) over 48 hr. Based on the observed metabolic disruptions, exposure to MeOH and DMSO induced higher metabolic perturbations in amino acid levels and associated biochemical pathways in comparison to ACT and ACN exposures. However, as with mixtures, when co-solvents are combined with the pollutants under investigation, there is a possibility for additive, synergistic, or antagonistic interactions. Hence, to examine the possible impairments in co-solvent and pollutant mixtures, ACT and ACN applied at 0.01% v/v were chosen to be tested with phenanthridine (PN). Daphnia magna exposure to PN dissolved in ACT had less disruptions; in contrast to PN prepared in ACN, which triggered a higher degree of antagonism in the D. magna metabolic profile. Consequently, exposing D. magna to ACT applied at 0.01% v/v resulted in the lowest metabolic perturbation in both parts of this study, suggesting that it is the least disruptive co-solvent for molecular-level exposure studies involving D. magna.
Collapse
Affiliation(s)
- Salwa Hajir
- Environmental NMR Centre and Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Karl J Jobst
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Sonya Kleywegt
- Technical Assessment and Standards Development Branch, Ontario Ministry of the Environment, Conservation and Parks, Toronto, Ontario, Canada
| | - André J Simpson
- Environmental NMR Centre and Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Myrna J Simpson
- Environmental NMR Centre and Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Göbel C, Niccolai R, de Groot MHP, Jayachandran J, Traets J, Kloosterman DJ, Gregoricchio S, Morris B, Kreft M, Song JY, Azarang L, Kasa E, Oskam N, de Groot D, Hoekman L, Bleijerveld OB, Kersten MJ, Aslam MA, van Leeuwen F, Jacobs H. Targeting DOT1L and EZH2 synergizes in breaking the germinal center identity of diffuse large B-cell lymphoma. Blood 2025; 145:1802-1813. [PMID: 39792929 DOI: 10.1182/blood.2024025500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/27/2024] [Accepted: 10/13/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Differentiation of antigen-activated B cells into proproliferative germinal center (GC) B cells depends on the activity of the transcription factors myelocytoma (MYC) and B-cell lymphoma 6 (BCL6), and the epigenetic writers disruptor of telomeric silencing 1-like (DOT1L) and enhancer of zeste homolog 2 (EZH2). GCB-like diffuse large B-cell lymphomas (GCB-DLBCLs) arise from GCB cells and closely resemble their cell of origin. Given the dependency of GCB cells on DOT1L and EZH2, we investigated the role of these epigenetic regulators in GCB-DLBCLs and observed that GCB-DLBCLs synergistically depend on the combined activity of DOT1L and EZH2. Mechanistically, inhibiting both enzymes led to enhanced derepression of polycomb repressive complex 2 target genes compared with EZH2 single treatment, along with the upregulation of BCL6 target genes and suppression of MYC target genes. The sum of all these alterations results in a "cell identity crisis," wherein GCB-DLBCLs lose their proproliferative GC identity and partially undergo plasma cell differentiation, a state associated with poor survival. In support of this model, combined epidrugging of DOT1L and EZH2 prohibited the outgrowth of human GCB-DLBCL xenografts in vivo. We conclude that the malignant behavior of GCB-DLBCLs strongly depends on DOT1L and EZH2 and that combined targeting of both epigenetic writers may provide an alternative differentiation-based treatment modality for GCB-DLBCL.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Germinal Center/pathology
- Germinal Center/metabolism
- Germinal Center/drug effects
- Animals
- Mice
- Histone-Lysine N-Methyltransferase/antagonists & inhibitors
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Differentiation
- Epigenesis, Genetic
Collapse
Affiliation(s)
- Camiel Göbel
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachele Niccolai
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jayashree Jayachandran
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joleen Traets
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ben Morris
- Robotics and Screening Center, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maaike Kreft
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leyla Azarang
- Biostatistics Center, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eirini Kasa
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nienke Oskam
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Daniel de Groot
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Liesbeth Hoekman
- Mass Spectrometry/Proteomics Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Onno B Bleijerveld
- Mass Spectrometry/Proteomics Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marie José Kersten
- Department of Hematology, Amsterdam University Medical Center (location University of Amsterdam), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Muhammad A Aslam
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Fred van Leeuwen
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center (location University of Amsterdam), Amsterdam, The Netherlands
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Hu P, Su L, Wang Y, Chen Y, Tian X, Qian Y. Targeting Liver Fibrosis with Nanoparticle Technology: The Dual-Drug Strategy for Hepatic Stellate Cell Activation Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40238180 DOI: 10.1021/acsami.5c02796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Hepatic stellate cells (HSCs) are pivotal in the pathogenesis and progression of liver fibrosis. Their activation is characterized by increased expression of integrin receptor αvβ3 and elevated intracellular oxidative stress, leading to extracellular matrix deposition. To address these challenges, we developed a nanotechnology-driven drug delivery system for the targeted transport of curcumin (CUR) and dihydromyricetin (DHM), two potential antifibrotic drugs with anti-inflammatory and antioxidant properties, into activated HSCs. Our results demonstrated that intravenously administered cyclo-RGD peptide (cRGDfk)-modified drug-loaded nanoparticles (NPs) effectively targeted fibrotic liver tissues, particularly activated HSCs. These drug-loaded NPs inhibited HSC activation and migration, induced apoptosis in activated HSCs, and downregulated α-SMA expression. In a carbon tetrachloride (CCl4)-induced liver fibrosis model, the NPs exhibited significant antifibrotic effects and reduced the number of Ly6Chi monocyte-derived macrophages in the liver. These findings suggest that cRGDfk-modified NPs carrying CUR and DHM have potential clinical applications in liver fibrosis therapy.
Collapse
Affiliation(s)
- Panyi Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, Anhui, People's Republic of China
| | - Liping Su
- State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan Province, People's Republic of China
| | - Yongchao Wang
- State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan Province, People's Republic of China
| | - Yongqiang Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, Anhui, People's Republic of China
| | - Xiaohe Tian
- State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan Province, People's Republic of China
| | - Yeben Qian
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230001, Anhui, People's Republic of China
| |
Collapse
|
8
|
Huo J, Nie K, Yang T, Zhang S, Zhu Z, Peng X, Zhang Y. Network pharmacology combined with transcriptomics reveals that Ganoderma lucidum spore and Sanghuangporus vaninii compound extract exerts anti-colorectal cancer effects via CYP24A1-mediated VDR pathway and TERT-mediated Wnt signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025:119820. [PMID: 40245966 DOI: 10.1016/j.jep.2025.119820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As traditional medicinal fungi, Ganoderma lucidum and Sanghuangporus vaninii are widely used in the treatment of tumor related diseases and cancer adjuvant therapy with potent anticancer effects. However, the anticancer effect and mechanism of action of their compound extract remain unclear. AIM OF THE STUDY To investigate the anticancer effect of Ganoderma lucidum and Sanghuangporus vaninii compound extract and explore the underlying mechanism. MATERIALS AND METHODS First, MTT assay was performed to investigate the effect of 8 different extracts on tumor cell viability. Moreover, the synergistic effect of Ganoderma lucidum spore and Sanghuangporus vaninii was evaluated by Chou-Talalay method. Subsequently, the fractional extractions were conducted to further isolate anti-tumor active components. Next, network pharmacology combined with transcriptomics was used to explore the potential mechanisms underlying the anticancer effect of compound extract. Finally, the mechanism of action was verified using in vitro and in vivo models. RESULTS Among all 8 extracts, Ganoderma lucidum spore and Sanghuangporus vaninii compound ethanol extract (GSEE) showed the most significant cell viability inhibitory effect on cancer cells, especially colorectal cancer (CRC) cells, which was even better than combination of Sanghuangporus vaninii ethanol extract (SVEE) and Ganoderma lucidum spore ethanol extract (GLEE). The ethyl acetate fraction of GSEE (GSEAE) was screened as the anti-tumor active fraction of GSEE and could suppress CRC proliferation in vitro and in vivo. The CYP24A1-mediated Vitamin D receptor (VDR) pathway and TERT-mediated Wnt signaling pathway were identified as the main mechanisms of GSEAE against CRC. Multiple CRC models confirmed that GSEAE suppressed CRC metastasis, arrested cell cycle and induced mitochondrial apoptosis of CRC cells via VDR pathway and Wnt signaling pathway. CONCLUSIONS Collectively, our data suggest that compound extract GSEAE exerts anti-CRC effects via CYP24A1-mediated VDR pathway and TERT-mediated Wnt signaling pathway.
Collapse
Affiliation(s)
- Jian Huo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Kun Nie
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Tianfeng Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Suyu Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Xiuhong Peng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering Xi'an 710061 P. R. China.
| |
Collapse
|
9
|
Lee H, Lee MR, Fan TM, Hergenrother PJ. PAC-1 Synergizes with Sunitinib to Enhance Cell Death in Pancreatic Neuroendocrine Tumors. ACS Pharmacol Transl Sci 2025; 8:1140-1151. [PMID: 40242587 PMCID: PMC11997889 DOI: 10.1021/acsptsci.5c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Pancreatic neuroendocrine tumors (PNETs) are rare tumors that are often diagnosed at advanced or metastatic stages, resulting in a poor prognosis. Sunitinib is an approved therapy for treatment of patients with PNETs, but low response rates and resistance have limited its impact, with autophagy and sunitinib sequestration in the lysosome identified as key resistance mechanisms. Here, we show that the combination of sunitinib with the procaspase-3 activator PAC-1 enhances PNET cell death in cell culture and in vivo in a xenograft tumor model. PAC-1 treatment enlarges lysosomes, resulting in partial lysosomal membrane permeabilization and blocking of autophagosome-lysosome fusion. These alterations lead to increased accumulation of autophagic structures, blocking autophagic flux, and a changed distribution of sunitinib from the lysosome to the cytosol. Our data show that PAC-1 modulates sunitinib-induced autophagy and blocks lysosomal trapping, potentiating sunitinib activity and increasing death of cancer cells. As both drugs are well-tolerated in patients, the data suggest evaluation of the PAC-1/sunitinib combination in a clinical trial of patients with PNET.
Collapse
Affiliation(s)
- Hyang
Yeon Lee
- Department
of Chemistry, University of Illinois at
Urbana−Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Myung Ryul Lee
- Department
of Chemistry, University of Illinois at
Urbana−Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
- Department
of Veterinary Clinical Medicine, University
of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department
of Chemistry, University of Illinois at
Urbana−Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
10
|
Ma X, Li H, Li Y, Xie X, Wang Y, Wang M, Peng X. Potential Antidiabetic Activity of Nordihydroguaiaretic Acid: An Insight into Its Inhibitory Mechanisms on Carbohydrate-Hydrolyzing Enzymes, the Binding Behaviors with Enzymes, and In Vivo Antihyperglycemic Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8290-8304. [PMID: 40152424 DOI: 10.1021/acs.jafc.4c11307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The inhibitory mechanisms and binding behaviors of nordihydroguaiaretic acid (NDGA) to α-glucosidase/α-amylase were investigated by in vitro multispectroscopic methods and in silico modeling technique. The results demonstrated that NDGA reversibly and uncompetitively inhibited α-glucosidase, exhibiting stronger inhibition than acarbose, while it displayed noncompetitive inhibition against α-amylase. Additionally, NDGA could spontaneously bind to α-glucosidase/α-amylase mainly through hydrogen bonds and hydrophobic forces, thus altering the spatial structure of enzymes and reducing their catalytic activity. The presence of crowding reagents/polysaccharides/undigested milk proteins would decrease the inhibitory ability of NDGA, whereas fatty acids exhibited the opposite phenomenon on α-glucosidase. Furthermore, the antidiabetic activity of NDGA in vivo was evaluated using the diabetic Drosophila model induced by a high-sugar diet. It was found that NDGA significantly reduced the glucose levels of diabetic Drosophila. These findings suggested that NDGA was a potential inhibitor of α-glucosidase/α-amylase and could be used as a nutritional adjuvant to prevent diabetes.
Collapse
Affiliation(s)
- Xiangzhao Ma
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Huan Li
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Mengfan Wang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xin Peng
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
11
|
Nimal S, Kumbhar N, Pote MS, Bankar R, Shaikh M, Gacche R. Reversal of epithelial to mesenchymal transition in triple negative breast cancer through epigenetic modulations by dietary flavonoid Galangin and its combination with SAHA. Cell Commun Signal 2025; 23:163. [PMID: 40176095 PMCID: PMC11967073 DOI: 10.1186/s12964-025-02174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND TNBC is an aggressive metastatic cancer that poses considerable treatment challenges because of its acquired drug resistance towards the existing targeted and hormonal therapies. The epigenetic modulation including HDACs triggers the EMT in TNBC which produces a more aggressive tumor phenotype. Chemotherapy and radiotherapy cause severe side effects which make treatment complex and challenging. To avoid these serious side effects and boost the effectiveness of current anti-cancer medications, plant flavonoids have been investigated. AIM OF THE STUDY The present investigation is aimed to understand the role of dietary flavonoid Gal in the modulation of epigenetic regulators such as HDACs and HATs and their impact on the reversal of the EMT process in TNBCs. METHODOLOGY Here, we have examined the anti-TNBC potential of Gal alone and in combination with SAHA by performing series of in vitro cell culture assays such MTT, migration and invasion, cell cycle regulation, ROS generation & mitochondrial dysfunction, nuclear fragmentation & apoptosis induction etc. The expression profiles of epigenetic regulators, apoptosis regulating proteins, and EMT markers were analysed by performing transcriptomic and proteomic studies. The in vivo efficacy of Gal was studied using BALB/c mice xenograft model studies. RESULTS At IC50 = 50 µM/mL, Gal significantly inhibited the cell proliferation, migration, and invasion, arrested cell cycle at sub G0/G1 phases, generated ROS, reduced MMP and induced apoptosis in MDA-MB-231. Transcriptomic, proteomic, and calorimetric analysis revealed that Gal has potential to downregulate the expression of HDAC1/HDAC3 and elevate the expression levels of HAT. Gal also modulated the process of EMT by downregulating the mesenchymal markers and upregulating the epithelial marker. The synergistic mechanism of Gal and SAHA against the TNBCs was elucidated by understanding the expression levels of epigenetic regulators & EMT markers. Interestingly, Gal increased the expression of tumour suppressor protein pTEN and suppressed the expression of AKT, PI3K, and mTOR proteins involved in the cancer proliferation pathway. Gal also demonstrated impressive antitumor effect under in vivo settings. CONCLUSION In-vitro and In vivo studies confirmed Gal's potent anticancer efficacy and highlighted its potential as a promising therapeutic agent that possibly can be used with conventional chemotherapy against TNBC.
Collapse
Affiliation(s)
- Snehal Nimal
- Department of Biotechnology, Savitribai Phule Pune University Pune, Pune, Maharashtra (MS), 411007, India
| | - Navanath Kumbhar
- Department of Biotechnology, Savitribai Phule Pune University Pune, Pune, Maharashtra (MS), 411007, India
| | - Manasi S Pote
- Department of Biotechnology, Savitribai Phule Pune University Pune, Pune, Maharashtra (MS), 411007, India
| | - Rahul Bankar
- National Centre for Cell Science, Pune, 411007, India
| | | | - Rajesh Gacche
- Department of Biotechnology, Savitribai Phule Pune University Pune, Pune, Maharashtra (MS), 411007, India.
- Department of Biotechnology, Savitribai Phule Pune University (SPPU), Pune, 411007, India.
| |
Collapse
|
12
|
Zhang T, Li H, Lin C, Wang X. Quantitative analysis of drug-drug interactions among active components of Xuebijing in inhibiting LPS-induced TLR4 signaling and NO production. Sci Rep 2025; 15:11103. [PMID: 40169827 PMCID: PMC11961611 DOI: 10.1038/s41598-025-95994-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/25/2025] [Indexed: 04/03/2025] Open
Abstract
Despite the long history of Traditional Chinese Medicine (TCM) in disease treatment, the underlying "Jun-Chen-Zuo-Shi" principle remains largely unexplored. To address this gap, it is essential to elucidate the interactions between active substances in TCM through quantitative molecular and cellular pharmacology. The Chou-Talalay method is particularly effective for investigating drug combinations, making it highly relevant for TCM formulas. This study employed the Chou-Talalay method to explore the drug-drug interactions in Xuebijing (XBJ), a TCM formula used for treating sepsis. The aim was to elucidate the "Jun-Chen-Zuo-Shi" principle by investigating the interactions of the main active substances in XBJ: danshensu and salvianolic acid B (from Radix Salviae Miltiorrhizae), senkyunolide A (from Rhizoma Chuanxiong), ligustilide (from Radix Angelicae Sinensis), safflower yellow and hydroxysafflor yellow A (from Flos Carthami), and paeoniflorin (from Radix Paeoniae Rubra). We quantitatively analyzed their TLR4 antagonistic activities and used the combination index (CI) to quantify their interactions, revealing synergism (CI < 1), additive effects (CI = 1), and antagonism (CI > 1). The results show these agents inhibit nitric oxide (NO) production, with some combinations demonstrating synergistic effects at certain concentrations, while others present antagonistic effects. Understanding these interactions provides a scientific foundation for optimizing TCM formulations, enhancing quality control, efficacy, and safety.
Collapse
Affiliation(s)
- Tianshu Zhang
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
| | - Xiaohui Wang
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China.
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, Jilin, China.
| |
Collapse
|
13
|
Xie DM, Li ZY, Ren BK, Gong R, Yang D, Huang S. Tanshinone II A Facilitates Chemosensitivity of Osteosarcoma Cells to Cisplatin via Activation of p38 MAPK Pathway. Chin J Integr Med 2025; 31:326-335. [PMID: 39499413 DOI: 10.1007/s11655-024-4118-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 11/07/2024]
Abstract
OBJECTIVE To examine the mechanism of action of tanshinone II A (Tan II A) in promoting chemosensitization of osteosarcoma cells to cisplatin (DDP). METHODS The effects of different concentrations of Tan II A (0-80 µ mol/L) and DDP (0-2 µ mol/L) on the proliferation of osteosarcoma cell lines (U2R, U2OS, 143B, and HOS) at different times were examined using the cell counting kit-8 and colony formation assays. Migration and invasion of U2R and U2OS cells were detected after 24 h treatment with 30 µ mol/L Tan II A, 0.5 µ mol/L DDP alone, and a combination of 10 µ mol/L Tan II A and 0.25 µ mol/L DDP using the transwell assay. After 48 h of treatment of U2R and U2OS cells with predetermined concentrations of each group of drugs, the cell cycle was analyzed using a cell cycle detection kit and flow cytometry. After 48 h treatment, apoptosis of U2R and U2OS cells was detected using annexin V-FITC apoptosis detection kit and flow cytometry. U2R cells were inoculated into the unilateral axilla of nude mice and then the mice were randomly divided into 4 groups of 6 nude mice each. The 4 groups were treated with equal volume of Tan II A (15 mg/kg), DDP (3 mg/kg), Tan II A (7.5 mg/kg) + DDP (1.5 mg/kg), and normal saline, respectively. The body weight of the nude mice was weighed, and the tumor volume and weight were measured. Cell-related gene and signaling pathway expression were detected by RNA sequencing and Kyoto Encyclopedia of Genes and Genomes pathway analysis. p38 MAPK signaling pathway proteins and apoptotic protein expressions were detected by Western blot. RESULTS In vitro studies have shown that Tan II A, DDP and the combination of Tan II A and DDP inhibit the proliferation, migration and invasion of osteosarcoma cells. The inhibitory effect was more pronounced in the Tan II A and DDP combined treatment group (P<0.05 or P<0.01). Osteosarcoma cells underwent significantly cell-cycle arrest and cell apoptosis by Tan II A-DDP combination treatment (P<0.05 or P<0.01). In vivo studies demonstrated that the Tan II A-DD combination treatment group significantly inhibited tumor growth compared to the Tan II A and DDP single drug group (P<0.01). Additionally, we found that the combination of Tan II A and DDP treatment enhanced the p38 MAPK signaling pathway. Western blot assays showed higher p-p38, cleaved caspase-3, and Bax and lower caspase-3, and Bcl-2 expressions with the combination of Tan II A and DDP treatment compared to the single drug treatment (P<0.01). CONCLUSION Tan II A synergizes with DDP by activating the p38/MAPK pathway to upregulate cleaved caspase-3 and Bax pro-apoptotic gene expressions, and downregulate caspase-3 and Bcl-2 inhibitory apoptotic gene expressions, thereby enhancing the chemosensitivity of osteosarcoma cells to DDP.
Collapse
Affiliation(s)
- Da-Ming Xie
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China
| | - Zhi-Yun Li
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Bing-Kai Ren
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Rui Gong
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, 330052, China
| | - Dong Yang
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China
| | - Sheng Huang
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China.
| |
Collapse
|
14
|
Frederick MI, Fyle E, Clouvel A, Abdesselam D, Hassan S. Targeting FEN1/EXO1 to enhance efficacy of PARP inhibition in triple-negative breast cancer. Transl Oncol 2025; 54:102337. [PMID: 40054125 PMCID: PMC11928819 DOI: 10.1016/j.tranon.2025.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. The only targeted therapeutic approach that has emerged for early TNBC patients with BRCA-mutations (BRCAMUT) are PARP inhibitors (PARPi). In combination, PARPi may benefit a larger cohort of TNBC patients. We used our previously identified 63-gene signature that was associated with PARPi response to identify candidate genes that could be therapeutic targets. We selected FEN1 for further investigation since its knockdown was associated with an increase in G2/M arrest, DNA damage, and apoptosis. We first tested LNT1, a FEN1/EXO1 inhibitor, in a panel of 10 TNBC cell lines. LNT1 sensitivity was identified predominantly in BRCA1-mutant/deficient cell lines. However, the combination of PARPi and LNT1 demonstrated a synergistic or additive effect in 7/10 cell lines, mainly in BRCA1/2 wild-type (BRCAWT) and BRCA2-mutant cell lines, with intrinsic and acquired resistance to PARPi. The greatest synergy was observed in a BRCA2-mutant cell line with acquired resistance to olaparib (HCC1395-OlaR), with a combination index value of 0.20. In the synergistic cell lines, BT549 (BRCAWT) and HCC1395-OlaR, the combination was associated with a rapid progression in DNA replication fork speed, an early and sustained increase in DNA damage in comparison to each of the single-agents. However, in the additive BRCA1/2 wild-type cell lines, MDAMB231 and HCC1806, the combination demonstrated a high DNA damage response that was largely driven by either talazoparib or LNT1. Therefore, targeting FEN1/EXO1 with PARPi is a promising targeted combination approach, particularly in the context of PARPi-resistant and BRCAWT TNBC.
Collapse
Affiliation(s)
- Mallory I Frederick
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Elicia Fyle
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Anna Clouvel
- Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Djihane Abdesselam
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada
| | - Saima Hassan
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3T5, Canada; Centre de Recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), l'Institut de Cancer de Montreal, Montreal, QC H2X0A9, Canada; Division of Surgical Oncology, Department of Surgery, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, QC H2X0C1, Canada.
| |
Collapse
|
15
|
Firat YY, Cicek B, Kara A, Ozturk NK, Ilgun S. Effects of Thyme, Cumin, and Sumac Extracts on Apoptosis and Paraptosis in Hepatocellular Carcinoma: Synergistic, Antagonistic, or Additive Properties. Food Sci Nutr 2025; 13:e70106. [PMID: 40129995 PMCID: PMC11931446 DOI: 10.1002/fsn3.70106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/05/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
This study evaluated the effect of single, double, and triple combined doses of sumac, thyme, and cumin extracts on apoptosis and paraptosis in the HepG2 cell line. The effect of thyme and cumin extracts was higher in proteins (mTOR, caspase-8, caspase-9, Bax and bcl-2) other than caspase-3 protein. The expression of caspase-3 protein was higher in the sumac extract-treated groups. The expression levels of GRP78/Bip and DDIT3/Chop proteins, which are indicators of paraptosis, did not exert a significant difference between the extracts. Even though their protein expression is different, according to MTT results, sumac and thyme extracts showed an additive effect, thyme and cumin extracts showed an antagonistic effect, sumac and cumin extracts showed a synergistic effect, and sumac, thyme, and cumin extracts showed a synergistic effect. Sumac, thyme, and cumin extracts induced cell death by causing apoptosis in HepG2 cells, and they may have a supportive impact on the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yagmur Yasar Firat
- Department of Nutrition Dietetic, Faculty of Health SciencesErciyes UniversityKayseriTürkiye
| | - Betul Cicek
- Department of Nutrition Dietetic, Faculty of Health SciencesErciyes UniversityKayseriTürkiye
| | - Ayca Kara
- Betül Ziya Eren Genom and Stem Cell CenterErciyes UniversityKayseriTürkiye
| | - Nurefsan Konyaligil Ozturk
- Department of Nutrition Dietetic, Faculty of Health SciencesBolu Abant İzzet Baysal UniversityBoluTürkiye
| | - Selen Ilgun
- Department of Nutrition Dietetic, Faculty of Health SciencesBolu Abant İzzet Baysal UniversityBoluTürkiye
- Department of Pharmacognosy, Faculty of PharmacyErciyes UniversityKayseriTürkiye
| |
Collapse
|
16
|
Yang Y, Xu Q, Gu W, Nan K, Chen S, Wang S, Zhang J, Zhao Q. Oxidative stress-augmented Cu-doped hollow mesoporous carbon nanozyme for photothermal/photodynamic synergistic therapy. J Colloid Interface Sci 2025; 683:910-925. [PMID: 39709766 DOI: 10.1016/j.jcis.2024.12.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Photodynamic therapy (PDT) has witnessed remarkable progress in recent years owing to its specific properties. Given that the antioxidation system of tumor microenvironment (TME) adversely affects treatment outcomes, powerful TME modulators can significantly resolve the limitation of PDT. Herein, we developed a PEG-modified Cu2+-doped hollow mesoporous carbon nanozyme (CHC-PEG) and loaded insoluble photosensitizer IR780 into its pores and cavities to construct the multifunctional nano-system IR780/CHCP. CHC-PEG nanozyme could perform photothermal therapy (PTT) effect and protect IR780 from aggregation-caused quenching (ACQ) effect, while exerting peroxidase (POD)-mimetic activity and the ability of consuming glutathione (GSH) to achieve oxidative stress-augmented PDT effect. When exposed to near-infrared (NIR) light, IR780 was stimulated to produce singlet oxygen (1O2) and CHC-PEG could increase the temperature of TME to exert stronger POD-mimetic activity for producing more hydroxyl radicals (OH), therefore the IR780/CHCP nano-system exhibited remarkable tumor growth inhibition. Benefited by the enhanced synergistic effect, IR780/CHCP exhibited remarkable in vivo tumor growth inhibition, with the tumor inhibition rate of 93 %, and had no significant effect on major organs. Above all, IR780/CHCP could resist the antioxidant system in TME to enhance the level of oxidative stress, thereby enabling effective anti-tumor therapy. This study introduced a novel strategy to effectively promote the synergistic PTT/PDT effect by the enhanced oxidative stress.
Collapse
Affiliation(s)
- Yuanqi Yang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Wei Gu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Kaisheng Nan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siyu Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
17
|
Muraoka H, Kazuno H, Hashimoto A, Sootome H, Ohkubo S. Pimitespib, an HSP90 Inhibitor, Enhances the Efficacy of PARP Inhibitors in PARP Inhibitor-Insensitive Breast Cancer Cells. Cancer Sci 2025. [PMID: 40167031 DOI: 10.1111/cas.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 02/18/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Heat shock protein 90 (HSP90) plays a crucial role in the maintenance of protein homeostasis in cancer cells. Inhibition of HSP90 is anticipated to exert anticancer activities by reducing levels of HSP90 client proteins. Pimitespib (TAS-116) has emerged as a potent ATP-competitive inhibitor of both HSP90α and β, demonstrating favorable therapeutic properties in preclinical models. Notably, pimitespib is the first HSP90 inhibitor approved for the treatment of advanced gastrointestinal stromal tumors in Japan. Poly(ADP-ribose) polymerase (PARP) inhibitors target cancers susceptible to the homologous recombination (HR) pathway and are used for treating various types of tumors, particularly those harboring defects in HR repair pathways within DNA damage repair (DDR) such as mutations in breast cancer genes 1 and 2 (BRCA1 and BRCA2, respectively). However, PARP inhibitors have shown limited efficacy in HR-proficient tumors, and the development of resistance to PARP inhibitors via restoration of DDR systems poses a significant challenge. In this study, we explored the potential of pimitespib to enhance PARP inhibitor activity. In PARP inhibitor-insensitive breast cancer cell lines, pimitespib impaired HR pathway function by promoting the proteasome-mediated degradation of proteins involved in HR, such as BRCA1, BRCA2, and Rad51 homologous 1 (RAD51). Consequently, pimitespib enhanced antitumor activity and DNA damage induced by PARP inhibitors in vitro. In human breast cancer xenograft mouse models, pimitespib downregulated RAD51 proteins and augmented the antitumor effects of PARP inhibitors. These findings highlight the potential of pimitespib as a therapeutic agent in combination with PARP inhibitors to treat PARP inhibitor-insensitive cancers.
Collapse
Affiliation(s)
- Hiromi Muraoka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hiromi Kazuno
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Akihiro Hashimoto
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Hiroshi Sootome
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co., Ltd., Tsukuba, Ibaraki, Japan
| |
Collapse
|
18
|
Chaudhary A, Kumar A, Swain N, Chaudhary K, Sonker H, Dewan S, Patil RA, Singh RG. Endocytic Uptake of Self-Assembled Iridium(III) Nanoaggregates for Holistic Treatment of Metastatic 3D Triple-Negative Breast Tumor Spheroids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406809. [PMID: 39607393 DOI: 10.1002/smll.202406809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Triple-negative breast cancer (TNBC) presents a formidable challenge due to its aggressive behavior and limited array of treatment options available. This study focuses on employing nanoaggregate material of organometallic Ir(III) complexes for treating TNBC cell line MDA-MB-231. In this approach, Ir(III) complexes with enhanced cellular permeability are strategically designed and achieved through the incorporation of COOMe groups into their structure. The lead compound, IrL1, exhibits promiscuous nanoscale aggregation in RPMI cell culture media, characterized by a stable hydrodynamic effective diameter ranging from 190 to 202 nm over 48 h. With excellent photo-responsive contrast-enhanced cell imaging properties IrL1 exhibits an outstanding IC50, 48h value of 36.05± 0.03 nm when irradiated with 390 nm light in MDA-MB-231 (IC50, 48 h of Cisplatin is 5.29 µµ). In cell, investigation confirms that IrL1 nanoaggregates internalization via energy-dependent endocytosis undergo ferroptosis and ROS mediated cell death in MDA-MB-231 cells. Further, these in vivo studies using NOD-SCID mice confirmed that IrL1 exhibits a tendency to ablate tumors inoculated in mice models at therapeutically relevant doses. Thus, this comprehensive approach holds promise for expanding the repertoire of organometallic Ir(III) nanoaggregates with adaptable characteristics, thereby advancing their clinical utility of nanomedicine in the holistic treatment of metastatic 3D triple-negative breast tumor spheroids.
Collapse
Affiliation(s)
| | - Ashwini Kumar
- Department of Chemistry, IIT Kanpur, Kanpur, UP, 208016, India
| | - Nikhil Swain
- Department of Chemistry, IIT Kanpur, Kanpur, UP, 208016, India
| | - Kajal Chaudhary
- Department of Chemistry, IIT Kanpur, Kanpur, UP, 208016, India
| | - Himanshu Sonker
- Department of Chemistry, IIT Kanpur, Kanpur, UP, 208016, India
| | - Sayari Dewan
- Department of Chemistry, IIT Kanpur, Kanpur, UP, 208016, India
| | | | | |
Collapse
|
19
|
Rivera D, Bouras A, Mattioli M, Anastasiadou M, Pacentra AC, Pelcher O, Koziel C, Schupper AJ, Chanenchuk T, Carlton H, Ivkov R, Hadjipanayis CG. Magnetic hyperthermia therapy enhances the chemoradiosensitivity of glioblastoma. Sci Rep 2025; 15:10532. [PMID: 40148452 PMCID: PMC11950323 DOI: 10.1038/s41598-025-95544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer and is resistant to standard-of-care chemoradiation therapy (CRT). Magnetic hyperthermia therapy (MHT) exposes magnetic iron oxide nanoparticles (MIONPs) to an alternating magnetic field (AMF) to generate local hyperthermia. This study evaluated MHT-mediated enhancement of CRT in preclinical GBM models. Cell viability and apoptosis were assessed in GBM cell lines after water bath heating with radiation and/or temozolomide. Heating efficiency of MIONPs after intracranial delivery was measured in healthy mice. MHT with CRT was performed in syngeneic and patient-derived xenograft (PDX) GBM tumors. Tissue sections were analyzed for γ-H2AX, HSP90, CD4 + T cells, and microglial cells. Tumor burden and survival were assessed. Hyperthermia with radiation and temozolomide significantly reduced cell viability and increased apoptosis. Hyperthermia predominantly exhibited additive to synergistic interactions with both treatment modalities and reduced doses needed for tumor cell growth inhibition. In vivo, MHT with CRT decreased tumor burden and increased survival in PDX and syngeneic models. Immunohistochemistry showed increased γ-H2AX, HSP90, microglial activation, and CD4 + T cells after MHT in combination with CRT. Overall, adjuvant hyperthermia enhances CRT efficacy in GBM cells, with MHT improving survival outcomes in rodents. Sufficient intracranial heating and MIONP retention for repeated treatments was achieved, supporting further clinical translation.
Collapse
Affiliation(s)
- Daniel Rivera
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Milena Mattioli
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Chiara Pacentra
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Olivia Pelcher
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Corrine Koziel
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Tori Chanenchuk
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Constantinos G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
20
|
Devi ST, Kshetrimayum V, Heisnam R, Akula SJ, Radhakrishnanand P, Mukherjee PK, Singh KB, Sharma N. Investigating the impact of Terminalia chebula, an underutilized functional fruit, on oral squamous cell carcinoma: Exploring cell death mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119482. [PMID: 39938761 DOI: 10.1016/j.jep.2025.119482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
ETHNOPHARMALOGICAL RELEVANCE Terminalia chebula, known for its extensive use in traditional medicinal practices among indigenous cultures, is recognized for its effectiveness in treating various oral disorders. Healers in India and China utilize the ripe fruits of T. chebula to prevent and manage conditions such as dental cavities, gingivitis, bleeding gums and stomatitis. The fruits have also been traditionally used in Ayurvedic and Siddha medicines for treatment of various diseases including anticancer properties. It is also an important component of Tibetan traditional medicine used for the treatment of cancer. Studies have demonstrated the efficacy of T. chebula against lung and colon carcinoma. AIM OF THE STUDY Despite its historical significance in oral health, the potential of T. chebula against oral cancer has not been explored, warranting further investigation into its bioactive properties. This study aims to explore the therapeutic potential of the hydroalcoholic extract of Terminalia chebula fruits and its fractions against oral squamous cell carcinoma (OSCC) using SCC9 cells focusing on their cytotoxicity, anti-proliferative effect and the synergistic action of its ethyl-acetate fraction with cisplatin (CP). Additionally it seeks to identify the bioactive phytoconstituents in EAF were identified using LC-ESI-QTOF-MS. MATERIALS AND METHODS Antioxidant activity of TYH and its fraction were assessed using DPPH and ABTS assays. Total phenolic (TPC) and total flavonoid content (TFC) were quantified via Folin-ciocalteau and alluminium chloride assays respectively. Cytotoxic and antiproliferative effects were assessed using MTT assay, clonogenic assay and cell migration assay. Apoptosis in EAF treated SCC9 cells was analysed by using DAPI, Giemsa staining and flow cytometry using Annexin V-FITC/PI apoptosis detection kit. Intracellular reactive oxygen species (ROS) was assessed using H2DCFDA, western blotting examined expression of apoptosis related proteins in SCC9 cells. Combinational effect of EAF with cisplatin (CP) was also assessed and phytochemical constituents of EAF were analysed using LC-ESI-QTOF-MS. RESULTS The ethyl acetate fraction (EAF) showed the highest antioxidant activity (IC50 value of 8.16 ± 0.59 μg/mL and 4.99 ± 0.82 μg/mL in DPPH and ABTS assays respectively) which reciprocated with a high TPC and TFC (528.46 ± 2.59 mgGAE/g and 49.10 ± 1.61 mgQE/g dry weight of the extract respectively) content. EAF significantly reduced cell viability with an IC50 value of 86.73 ± 0.55 μg/mL, resulted in dose dependent cell death, and prevented the proliferation and migration in SCC9 cells. Further Annexin V-PI based flow cytometric analysis and caspase-3/7 enzyme activity assay confirmed the apoptotic effect of EAF in SCC9 cells. Intrinsic pathway of apoptosis post treatment with EAF was confirmed by western blotting with marker proteins, Bax, Bcl-2, Mcl-1, cleaved caspase, procaspase and PARP. A combinatorial study of EAF with the standard drug cisplatin also indicated a synergistic effect of the fraction in cisplatin treated cells with a CI value of 0.67571. LC-ESI-QTOF-MS led to identification of the presence of phenolics and gallotannins with anticancer properties in EAF. CONCLUSION This study demonstrates the potential of the hydroalcoholic extract of Terminalia chebula fruits (TYH), especially its ethyl acetate fraction (EAF), as a therapeutic agent against oral squamous cell carcinoma (OSCC).
Collapse
Affiliation(s)
- Soibam Thoithoisana Devi
- Institute of Bioresources and Sustainable Development (An Autonomous Institute Under the Department of Biotechnology, Govt. of India) Takyelpat, Imphal- 795001, Manipur, India; Department of Zoology, Manipur University (MU), Imphal- 795003, Manipur, India
| | - Vimi Kshetrimayum
- Institute of Bioresources and Sustainable Development (An Autonomous Institute Under the Department of Biotechnology, Govt. of India) Takyelpat, Imphal- 795001, Manipur, India; School of Biotechnology Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University Bhubaneshwar, Odisha-751024, India
| | - Rameshwari Heisnam
- Institute of Bioresources and Sustainable Development (An Autonomous Institute Under the Department of Biotechnology, Govt. of India) Takyelpat, Imphal- 795001, Manipur, India; School of Biotechnology Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University Bhubaneshwar, Odisha-751024, India
| | - Sai Jyothi Akula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, 781101, India
| | - Pullapanthula Radhakrishnanand
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, 781101, India
| | - Pulok K Mukherjee
- Institute of Bioresources and Sustainable Development (An Autonomous Institute Under the Department of Biotechnology, Govt. of India) Takyelpat, Imphal- 795001, Manipur, India
| | | | - Nanaocha Sharma
- Institute of Bioresources and Sustainable Development (An Autonomous Institute Under the Department of Biotechnology, Govt. of India) Takyelpat, Imphal- 795001, Manipur, India.
| |
Collapse
|
21
|
Li M, Li C, Tao T, Li W, Kang M, Fu P, Liu X, Quan W, Zhu M, Liu Z. Study on the synergistical effects of characteristic compounds in Osmanthus black tea against xanthine oxidase based on multispectral analysis combined with in silico studies. Int J Biol Macromol 2025; 308:142479. [PMID: 40154674 DOI: 10.1016/j.ijbiomac.2025.142479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
With the worldwide prevalence of hyperuricemia (HUA), safe and effective natural xanthine oxidase (XOD) inhibitors are in need. This research was aimed to reveal the promising XOD inhibitors in Osmanthus black tea. Here, the combination index (CI) values for TF3 and acteoside were below 0.9, confirming the synergistic inhibitory effects on XOD. Further research confirmed that TF3-acteoside was stronger in inducing static quenching of XOD fluorescence than TF3 or acteoside. Besides, the secondary structure of XOD was significant changed by TF3-acteoside, specifically a decrease in the content of α-Helix and random coil, accompanied by an increase in β-Sheet and β-Turn contents, ultimately its structural stability and rigidity was enhanced. Molecular docking and molecular dynamics simulation analysis verified that TF3-acteoside stably bound to XOD by multiple hydrogen bonds. This study will lay important theoretical basis for the advancement of novel XOD inhibitors and the application of osmanthus black tea in lowering uric acid (UA).
Collapse
Affiliation(s)
- Maiquan Li
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China
| | - Cui Li
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Tiantian Tao
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Wenlan Li
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Manjun Kang
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Peitao Fu
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Xia Liu
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Wei Quan
- College of Food Science and Technology, Hunan Provincial Key Laboratory of Food Science and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Mingzhi Zhu
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, College of Horticulture, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
22
|
Flörkemeier I, Hotze HL, Heyne AL, Hildebrandt J, Weimer JP, Hedemann N, Rogmans C, Holthaus D, Siebert FA, Hirt M, Polten R, Morgan M, Klapdor R, Schambach A, Dempfle A, Maass N, van Mackelenbergh MT, Clement B, Bauerschlag DO. Dual Topoisomerase Inhibitor Is Highly Potent and Improves Antitumor Response to Radiotherapy in Cervical Carcinoma. Int J Mol Sci 2025; 26:2829. [PMID: 40243435 DOI: 10.3390/ijms26072829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Despite advances in vaccination and early detection, the total number of cases and deaths from cervical cancer has risen steadily in recent decades, making it the fourth most common type of cancer in women worldwide. Low-income countries in particular struggle with limited resources and treatment limitations for cervical cancer. Thus, effective medicines that are simple to manufacture are needed. The newly developed dual topoisomerase inhibitor P8-D6, with its outstanding ability to induce apoptosis, could be a promising option. In this study, the efficacy of P8-D6 in combination with radiochemotherapy against cervical carcinoma was investigated in established cell lines and in a translational approach in ex vivo patient cells by measuring the cytotoxicity, cell viability and caspase activity in vitro in 2D and 3D cell cultures. Treatment with P8-D6 resulted in significantly greater cytotoxicity and apoptosis induction compared to standard therapeutic cisplatin in both 2D and 3D cell cultures. Specifically, a considerably stronger anti-proliferative effect was observed. The treatment also led to morphological changes and a loss of membrane integrity in the 3D spheroids. Radiotherapy also benefited greatly from P8-D6 treatment. In fact, P8-D6 was a more potent radiosensitizer than cisplatin. Simple synthesis, favorable physicochemical properties and high potency make P8-D6 a promising cervical cancer drug candidate.
Collapse
Affiliation(s)
- Inken Flörkemeier
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrecht University of Kiel, 24118 Kiel, Germany
- Priority Research Area Kiel Nano, Surface and Interface Sciences (KiNSIS), Kiel University, 24118 Kiel, Germany
| | - Hannah L Hotze
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Anna Lena Heyne
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrecht University of Kiel, 24118 Kiel, Germany
| | - Jonas Hildebrandt
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrecht University of Kiel, 24118 Kiel, Germany
| | - Jörg P Weimer
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Nina Hedemann
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Christoph Rogmans
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - David Holthaus
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Frank-André Siebert
- Clinic of Radiotherapy, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Markus Hirt
- Clinic of Radiotherapy, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Robert Polten
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Rüdiger Klapdor
- Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany
- Department of Gynecology and Obstetrics, Albertinen Hospital Hamburg, 22457 Hamburg, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Marion T van Mackelenbergh
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Bernd Clement
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrecht University of Kiel, 24118 Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynecology and Obstetrics, Kiel University and University Medical Center Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Gynecology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
23
|
Gbadegesin OD, Adesina SK. Gemcitabine-Doxorubicin Combination Polymer-Drug Conjugate Prepared by SPAAC Click Chemistry: In Vitro Characterization. Int J Mol Sci 2025; 26:2798. [PMID: 40141443 PMCID: PMC11943159 DOI: 10.3390/ijms26062798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Combination chemotherapy is preferred for the treatment of ovarian cancer (OC). Systemic toxicity, however, frequently limits the effectiveness of treatment. Polymer-drug conjugates (PDCs) containing synergistic combinations of chemotherapeutic drugs can be used to enhance therapeutic efficacy. We earlier reported the use of a strain-promoted [3 + 2] azide-alkyne cycloaddition (SPAAC)-mediated polymerization method for the preparation of single-drug PDCs. In this report, the polymerization method was used to prepare gemcitabine-doxorubicin combination PDC. The PDC had a high molecular weight (Mw 1360 kDa) and high drug loading (36.6% weight gemcitabine; 7.0% weight doxorubicin). It demonstrated cathepsin B-catalyzed drug release at pH 5.0 and good hydrolytic stability at pH 7.4. The combination index analysis of free gemcitabine and free doxorubicin showed a concentration-dependent synergism (combination index < 1) in OVCAR-3 OC cells. Compared to individual gemcitabine PDC (the concentration that inhibited 50% growth (IC50) > 50 µg/mL) and doxorubicin PDC (IC50 = 1.79 µg/mL), the combination PDC (IC50 = 0.99 µg/mL) showed greater cytotoxicity against OVCAR-3 cells and was less cytotoxic than the equivalent free drug combination (IC50 = 0.11 µg/mL). The gemcitabine-doxorubicin combination PDC is promising for targeted combination chemotherapy of OC.
Collapse
Affiliation(s)
| | - Simeon K. Adesina
- Department of and Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
24
|
Camila Marques DA, Cleni Mara Marzocchi M, Fabíola Attié DC, Natércia T, Cristina A, Felipe C, Nathália Cristina C, Maria Regina T. Antitumor and antiproliferative potential of crotoxin in triple negative breast tumors. Toxicon 2025; 258:108322. [PMID: 40120860 DOI: 10.1016/j.toxicon.2025.108322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Triple negative breast carcinoma represents around 15 % of breast cancer cases. Is a type of breast cancer with poor prognosis, because it has a high rate of recurrence, metastasis and death and can be resistant to therapy. The recommended treatment is the use of anthracyclines with taxanes, however, these medications have several side effects. Thus, there is a need for new therapeutic approaches. Crotoxin, the main toxin found in the venom of the South American rattlesnake Crotalus durissus terrificus, is a potent β-neurotoxin that has phospholipase A2 (PLA2) activity. It exhibits preferential cytotoxic activity against several types of tumor cells and is most cytotoxic to cell lines that express high levels of epidermal growth factor receptors. Considering this, in this study, we evaluated the biological mechanisms that trigger the antitumor effects of crotoxin in a cell line representing triple negative breast carcinoma (MDA-MB-231 tumor cells). Results demonstrated that crotoxin had anti-apoptotic, anti-autophagic and pro-necrotic actions. The pro-necrotic effect occurred through mechanisms of apoptosis evasion, autophagy inhibition and DNA damage. Therefore, this study represents an important milestone to better understand the effects and mechanisms of action of crotoxin in triple negative breast cancer.
Collapse
Affiliation(s)
- De Andrade Camila Marques
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil.
| | - Machado Cleni Mara Marzocchi
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil
| | - De Castro Fabíola Attié
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil
| | - Teixeira Natércia
- University of Porto, Faculty of Pharmacy, Department of Biological Sciences, Laboratory of Biochemistry, Porto, Portugal
| | - Amaral Cristina
- University of Porto, Faculty of Pharmacy, Department of Biological Sciences, Laboratory of Biochemistry, Porto, Portugal
| | - Campos Felipe
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil
| | - Canicoba Nathália Cristina
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil
| | - Torqueti Maria Regina
- University of São Paulo, Faculty of Pharmaceutical Sciences of Ribeirão Preto, Department of Clinical, Toxicological and Food Sciences, Ribeirão Preto, Brazil
| |
Collapse
|
25
|
do Prado-Souza LFL, Ferraz LS, Citrangulo Tortelli T, Ribeiro CAJ, do Amaral DT, Arruda DC, de Oliveira ÉA, Chammas R, Maria-Engler SS, Rodrigues T. Exploiting Paradoxical Activation of Oncogenic MAPK Signaling by Targeting Mitochondria to Sensitize NRAS Mutant-Melanoma to Vemurafenib. Int J Mol Sci 2025; 26:2675. [PMID: 40141318 PMCID: PMC11942190 DOI: 10.3390/ijms26062675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Vemurafenib is a BRAF (rapidly accelerated fibrosarcoma B-type)-targeted therapy used to treat patients with advanced, unresectable melanoma. It inhibits the MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-regulated kinase) pathway and tumor proliferation in BRAFV600E-mutated melanoma cells. Resistance to vemurafenib has been reported in melanoma patients due to secondary NRAS (neuroblastoma RAS viral oncogene homolog) mutations, which lead to paradoxical MAPK pathway activation and tumor proliferation. However, the impact of this paradoxical activation on mitochondrial dynamics and function in NRAS-mutated melanoma is unclear. Here, we investigated the effects of vemurafenib on NRASQ61R-mutated melanoma cells, focusing on mitochondrial dynamics and function. As expected, vemurafenib did not exhibit cytotoxicity in SK-MEL-147 NRASQ61R-mutated melanoma cells, even after 72 h of incubation. However, it significantly enhanced the MAPK/ERK signaling through paradoxical activation, accompanied by decreased expression of mitochondrial fusion proteins and activation of the fission protein DRP1 (dynamin-related protein 1), leading to small, rounded mitochondrial morphology. These observations were corroborated by transcriptome data obtained from NRAS-mutated melanoma patients, showing MFN1 (mitofusin 1) and OPA1 (optic atrophy 1) downregulation and DNM1L (DRP1 gene) upregulation. Interestingly, inhibition of mitochondrial fission with mdivi-1 or modulation of oxidative phosphorylation via respiratory chain inhibition or uncoupling significantly sensitized NRASQ61R-mutated melanoma cells to vemurafenib. Despite vemurafenib's low cytotoxicity in NRAS-mutated melanoma, targeting mitochondrial dynamics and/or oxidative phosphorylation may offer a promising strategy for combined therapy.
Collapse
Affiliation(s)
- Laura Francisca Leite do Prado-Souza
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, Sao Paulo 09210-580, Brazil; (L.F.L.d.P.-S.); (L.S.F.); (C.A.J.R.); (D.T.d.A.)
| | - Letícia Silva Ferraz
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, Sao Paulo 09210-580, Brazil; (L.F.L.d.P.-S.); (L.S.F.); (C.A.J.R.); (D.T.d.A.)
| | - Tharcísio Citrangulo Tortelli
- Center for Translational Research in Oncology (LIM24), Cancer Institute of the State of Sao Paulo (ICESP), Clinical Hospital of the University of Sao Paulo Medical School (HCFMUSP), Sao Paulo 01246-000, Brazil; (T.C.T.J.); (R.C.)
- Comprehensive Center for Precision Oncology, University of São Paulo, Sao Paulo 05508-220, Brazil
| | - César Augusto João Ribeiro
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, Sao Paulo 09210-580, Brazil; (L.F.L.d.P.-S.); (L.S.F.); (C.A.J.R.); (D.T.d.A.)
| | - Danilo Trabuco do Amaral
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, Sao Paulo 09210-580, Brazil; (L.F.L.d.P.-S.); (L.S.F.); (C.A.J.R.); (D.T.d.A.)
| | - Denise Costa Arruda
- Integrated Biotechnology Nucleus (NIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, Sao Paulo 08780-911, Brazil;
| | | | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Cancer Institute of the State of Sao Paulo (ICESP), Clinical Hospital of the University of Sao Paulo Medical School (HCFMUSP), Sao Paulo 01246-000, Brazil; (T.C.T.J.); (R.C.)
- Comprehensive Center for Precision Oncology, University of São Paulo, Sao Paulo 05508-220, Brazil
| | - Silvya Stuchi Maria-Engler
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-220, Brazil;
| | - Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo Andre, Sao Paulo 09210-580, Brazil; (L.F.L.d.P.-S.); (L.S.F.); (C.A.J.R.); (D.T.d.A.)
| |
Collapse
|
26
|
Cheng WM, Li PC, Nguyen MTB, Lin YT, Huang YT, Cheng TS, Nguyen TH, Tran TH, Huang TY, Hoang TH, Chen SY, Chu YC, Wu CW, Lee MF, Chiou YS, Liu HS, Hong YR, Chang PMH, Hu YF, Chang YC, Lai JM, Huang CYF. Repurposing pitavastatin and atorvastatin to overcome chemoresistance of metastatic colorectal cancer under high glucose conditions. Cancer Cell Int 2025; 25:79. [PMID: 40050889 PMCID: PMC11887183 DOI: 10.1186/s12935-025-03712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) poses a significant clinical challenge because of drug resistance, which can adversely impact patient outcomes. Recent research has shown that abnormalities within the tumor microenvironment, especially hyperglycemia, play a crucial role in promoting metastasis and chemoresistance, and thereby determine the overall prognosis of patients with advanced CRC. METHODS This study employs data mining and consensus molecular subtype (CMS) techniques to identify pitavastatin and atorvastatin as potential agents for targeting high glucose-induced drug resistance in advanced CRC cells. CRC cells maintained under either low or high glucose conditions were established and utilized to assess the cytotoxic effects of pitavastatin and atorvastatin, both with and without 5-fluorouracil (5-FU). CRC 3D spheroids cultured were also included to demonstrate the anti-drug resistance of pitavastatin and atorvastatin. RESULTS A bioinformatics analysis identified pitavastatin and atorvastatin as promising drug candidates. The CMS4 CRC cell line SW480 (SW480-HG) was established and cultured under high glucose conditions to simulate hyperglycemia-induced drug resistance and metastasis in CRC patients. Pitavastatin and atorvastatin could inhibit cell proliferation and 3D spheroid formation of CMS4 CRC cells under high glucose conditions. In addition, both pitavastatin and atorvastatin can synergistically promote the 5-FU-mediated cytotoxic effect and inhibit the growth of 5-FU-resistant CRC cells. Mechanistically, pitavastatin and atorvastatin can induce apoptosis and synergistically promote the 5-FU-mediated cytotoxic effect by activating autophagy, as well as the PERK/ATF4/CHOP signaling pathway while decreasing YAP expression. CONCLUSION This study highlights the biomarker-guided precision medicine strategy for drug repurposing. Pitavastatin and atorvastatin could be used to assist in the treatment of advanced CRC, particularly with CMS4 subtype CRC patients who also suffer from hyperglycemia. Pitavastatin, with an achievable dosage used for clinical interventions, is highly recommended for a novel CRC therapeutic strategy.
Collapse
Affiliation(s)
- Wei-Ming Cheng
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Urology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Division of Urology, Department of Surgery, Zhongxiao Branch, Taipei City Hospital, Taipei, 115, Taiwan
| | - Po-Chen Li
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Minh Tran-Binh Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Yu-Teng Lin
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Tai-Shan Cheng
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan
| | - Thi-Huong Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Biotechnology and Food Technology, Thai Nguyen University of Agriculture and Forestry, Thai Nguyen, Vietnam
| | - Thu-Ha Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Thu-Huyen Hoang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Sin-Yu Chen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu-Chieh Chu
- Taipei First Girls High School, Taipei, 110, Taiwan
| | - Chih-Wei Wu
- Taipei First Girls High School, Taipei, 110, Taiwan
| | - Ming-Fen Lee
- Department of Nutrition, China Medical University, Taichung, 406, Taiwan
| | - Yi-Shiou Chiou
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hsiao-Sheng Liu
- Medical Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- 13 M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Center for Cancer Research, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Yi-Ren Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Peter Mu-Hsin Chang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Yu-Feng Hu
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, 115, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jin-Mei Lai
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City, 242, Taiwan.
| | - Chi-Ying F Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
27
|
Rîmbu MC, Popescu L, Mihăilă M, Sandulovici RC, Cord D, Mihăilescu CM, Gălățanu ML, Panțuroiu M, Manea CE, Boldeiu A, Brîncoveanu O, Savin M, Grigoroiu A, Ungureanu FD, Amzoiu E, Popescu M, Truță E. Synergistic Effects of Green Nanoparticles on Antitumor Drug Efficacy in Hepatocellular Cancer. Biomedicines 2025; 13:641. [PMID: 40149616 PMCID: PMC11940350 DOI: 10.3390/biomedicines13030641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Cancer remains one of the leading causes of mortality worldwide. Despite significant advancements in treatment strategies and drug development, survival rates remain low and the adverse effects of conventional therapies severely impact patients' quality of life. This study evaluates the therapeutic potential of plant-derived extracts in hepatocellular carcinoma treatment, with a focus on minimizing side effects while enhancing efficacy. Methods: This research investigates the in vitro synergistic effect of silver bio-nanoparticles synthesized from Clematis vitalba, Melissa officinalis, and Taraxacum officinale extracts (Clematis vitalbae extractum-CVE, Melissae extractum-ME, Taraxaci extractum-TE) in combination with liver cancer drugs, sunitinib (SNTB) and imatinib (IMTB), on HepG2 (human hepatocellular carcinoma) and HUVEC (human umbilical vein endothelial) cell lines. The silver nanoparticles (AgNPs) were characterized using UV-Vis spectroscopy, dynamic light scattering (DLS), zeta potential analysis, and scanning electron microscopy (SEM). The antitumor effects were evaluated through cell viability assays after 24 and 48 h of exposure, with additional cytotoxicity tests on HUVEC cells. Results: Results indicated that Melissa officinalis-derived silver nanoparticles (ME AgNPs) and Clematis vitalba extract with silver nanoparticles (CVE AgNPs) significantly reduced HepG2 cell viability. Their efficacy improved when combined with conventional therapies (SNTB + ME AgNPs 1:1 vs. SNTB: 20.01% vs. 25.73%, p = 0.002; IMTB + ME AgNPs 1:1 vs. IMTB: 17.80% vs. 18.08%, p = 0.036; SNTB + CVE AgNPs 1:1 vs. SNTB: 18.73% vs. 25.73%, p = 0.000; SNTB + CVE AgNPs 1:2 vs. SNTB: 26.62% vs. 41.00%, p = 0.018; IMTB + CVE AgNPs 1:1 vs. IMTB: 12.99% vs. 18.08%, p = 0.001). Taraxacum extract exhibited similar cytotoxicity to its nanoparticle formulation but did not exceed the efficacy of the extract alone at 24 h. Selectivity index assessments confirmed that AgNPs-based formulations significantly improve cytotoxicity and selectivity to HepG2 cells. Among the tested extracts, CVE demonstrated the strongest antitumor effect, enhancing the efficacy of synthetic drugs (CI < 1). SNTB + TE AgNPs (5% EtOH) also demonstrated consistent synergy at high doses, while SNTB + CVE AgNPs provided broad-range synergy, making it suitable for dose-escalation strategies. Conclusions: These findings underscore the potential of nanoparticle-based formulations in combination therapies with targeted kinase inhibitors such as sunitinib and imatinib. Future research should focus on in vivo validation and clinical trials to confirm these findings.
Collapse
Affiliation(s)
- Mirela Claudia Rîmbu
- Medical Doctoral School, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (M.C.R.); (F.D.U.)
| | - Liliana Popescu
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| | - Mirela Mihăilă
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
- Ștefan S. Nicolau Institute of Virology, 285 Mihai Bravu Street, 030304 Bucharest, Romania
| | - Roxana Colette Sandulovici
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| | - Daniel Cord
- Medical Doctoral School, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (M.C.R.); (F.D.U.)
| | - Carmen-Marinela Mihăilescu
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
- National Institute for Research and Development in Microtechnologies (IMT Bucharest), 072996 Bucharest, Romania; (A.B.); (O.B.); (M.S.); (A.G.)
- Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Mona Luciana Gălățanu
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| | - Mariana Panțuroiu
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| | - Carmen-Elisabeta Manea
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
- Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Măgurele, Romania
| | - Adina Boldeiu
- National Institute for Research and Development in Microtechnologies (IMT Bucharest), 072996 Bucharest, Romania; (A.B.); (O.B.); (M.S.); (A.G.)
| | - Oana Brîncoveanu
- National Institute for Research and Development in Microtechnologies (IMT Bucharest), 072996 Bucharest, Romania; (A.B.); (O.B.); (M.S.); (A.G.)
| | - Mihaela Savin
- National Institute for Research and Development in Microtechnologies (IMT Bucharest), 072996 Bucharest, Romania; (A.B.); (O.B.); (M.S.); (A.G.)
| | - Alexandru Grigoroiu
- National Institute for Research and Development in Microtechnologies (IMT Bucharest), 072996 Bucharest, Romania; (A.B.); (O.B.); (M.S.); (A.G.)
| | - Florin Dan Ungureanu
- Medical Doctoral School, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (M.C.R.); (F.D.U.)
| | - Emilia Amzoiu
- Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Mariana Popescu
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| | - Elena Truță
- Faculty of Pharmacy, Titu Maiorescu University, Gheorghe Sincai Blv. 16, 040314 Bucharest, Romania; (L.P.); (M.M.); (R.C.S.); (M.L.G.); (M.P.); (C.-E.M.); (M.P.); (E.T.)
| |
Collapse
|
28
|
Lerksuthirat T, Prasopporn S, Wikiniyadhanee R, Chitphuk S, Stitchantrakul W, Owneium P, Jirawatnotai S, Dejsuphong D. DNA damage response mutations enhance the antitumor efficacy of ATR and PARP inhibitors in cholangiocarcinoma cell lines. Oncol Lett 2025; 29:128. [PMID: 39822940 PMCID: PMC11736248 DOI: 10.3892/ol.2025.14874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/28/2024] [Indexed: 01/19/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a biliary tract carcinoma that is challenging to treat due to its heterogeneity and limited treatment options. Genetic alterations in DNA damage response (DDR) pathways and homologous recombination (HR) defects are common in CCA. This has prompted interest in the use of ataxia telangiectasia and Rad3-related protein (ATR) and poly(ADP-ribose) polymerase (PARP) inhibitors to treat CCA. The present study investigated the impact of an ATR inhibitor and various PARP inhibitors, individually and in combination, on CCA cell lines with different DDR mutation profiles. DDR gene alterations in these cell lines were analyzed, and the responses of the cells to treatment with the PARP inhibitors olaparib, veliparib and talazoparib and/or the ATR inhibitor AZD6738 were evaluated. Assessments focused on cellular viability, clonogenic survival and the combination index, alongside changes in DNA damage assessed via the formation of micronuclei and γ-H2A histone family member X foci. The results revealed that the CCA cell lines with more DDR mutations exhibited greater sensitivity to single and combination treatments. Talazoparib was found to be the most potent PARP inhibitor in the CCA cell lines. The combination of AZD6738 and talazoparib demonstrated varying synergistic effects depending on the genetic background of the CCA cells, with greater efficacy in the cell lines less sensitive to single drug treatments. Mechanistically, this combination promoted the accumulation of DNA damage, including DNA double-strand breaks. Overall, the study underscores the importance of HR in CCA. It reveals an association between the extent of DDR mutations and the response to AZD6738 and PARP inhibitors in CCA, both as single agents and in combination. These findings highlight that the number of mutated genes influences variability in the drug response.
Collapse
Affiliation(s)
- Tassanee Lerksuthirat
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Sunisa Prasopporn
- Siriraj Center of Research for Excellence (SiCORE) for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Rakkreat Wikiniyadhanee
- Program in Translational Medicine, Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn 10540, Thailand
| | - Sermsiri Chitphuk
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Wasana Stitchantrakul
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Paravee Owneium
- Program in Translational Medicine, Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn 10540, Thailand
| | - Siwanon Jirawatnotai
- Siriraj Center of Research for Excellence (SiCORE) for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Donniphat Dejsuphong
- Program in Translational Medicine, Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn 10540, Thailand
| |
Collapse
|
29
|
Cuero-Amu K, Daniela Bonilla-Velásquez L, Vargas-Casanova Y, Lucía Leal-Castro A, Marcela Parra-Giraldo C, Giselle López-Sánchez A, Fierro-Medina R, García-Castañeda J, Rivera-Monroy Z. Linear and Polyvalent Peptides with Potent Antimicrobial Activity Against Sensitive and Multidrug-Resistant E. c oli Clinical Isolates. Chem Biodivers 2025; 22:e202401734. [PMID: 39486005 DOI: 10.1002/cbdv.202401734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 11/03/2024]
Abstract
Peptides containing the sequences 20RRWQWR25 and 20RRWQWRMKKLG30 derived from Bovine lactoferricin (LfcinB) were synthesized and their antibacterial effect against reference strains and sensitive and resistant clinical isolates of E. coli was evaluated. Tetra-branched multiple antigen peptide (MAP) ((RRWQWR)2-K-Ahx-C)2 exhibited significant antibacterial activity against sensitive, resistant, and multidrug-resistant clinical isolates of E. coli. Peptide 3: RRWQWR-Nal-KKLG; MIC=16 μM, 26[F]: (RRWQWRFKKLG)2-K-Ahx; MIC=15 μM, 17: (RRWQWRFK)2-K-Ahx; MIC=9 μM, and LfcinB (20-25)2: (RRWQWR)2-K-Ahx; MIC=11 μM exhibited the highest antibacterial activity against E. coli strains, with bactericidal effect and haemolytic effect at MIC less than 5 % and a therapeutic index >1. A synergistic effect of peptides 26[F] and 17 with ciprofloxacin (CIP) or ceftriaxone (CEF) was observed. Prolonged treatment of E. coli ATCC 25922 with sublethal concentrations of CIP induced resistance in this strain, whereas some peptides did not induce resistance. These peptides can be considered to be promising candidates for treating infections caused by resistant strains of E. coli.
Collapse
Affiliation(s)
- Kelin Cuero-Amu
- Instituto de Biotecnología, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | - Laura Daniela Bonilla-Velásquez
- Instituto de Biotecnología, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | | | - Aura Lucía Leal-Castro
- Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | | | - Amalia Giselle López-Sánchez
- Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | - Ricardo Fierro-Medina
- Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | - Javier García-Castañeda
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| | - Zuly Rivera-Monroy
- Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 45 No 26-85, 11321, Bogotá, Colombia
| |
Collapse
|
30
|
Das S, Thakur S, Cahais V, Virard F, Claeys L, Renard C, Cuenin C, Cros MP, Keïta S, Venuti A, Sirand C, Ghantous A, Herceg Z, Korenjak M, Zavadil J. Molecular and cell phenotype programs in oral epithelial cells directed by co-exposure to arsenic and smokeless tobacco. Biofactors 2025; 51:e70011. [PMID: 40056068 PMCID: PMC11962598 DOI: 10.1002/biof.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Chronic exposure to arsenic can lead to various health issues, including cancer. Concerns have been mounting about the enhancement of arsenic toxicity through co-exposure to various prevalent lifestyle habits. Smokeless tobacco (SLT) products are commonly consumed in South Asian countries, where their use frequently co-occurs with exposure to arsenic from contaminated groundwater. To decipher the in vitro molecular and cellular responses to arsenic and/or smokeless tobacco, we performed temporal multi-omics analysis of the transcriptome and DNA methylome remodeling in exposed hTERT-immortalized human normal oral keratinocytes (NOK), as well as arsenic and/or smokeless tobacco genotoxicity and mutagenicity investigations in NOK cells and in human p53 knock-in murine embryonic fibroblasts (Hupki MEF). RNAseq results from acute exposures of NOK cell to arsenic alone and in combination with smokeless tobacco extract revealed upregulation of genes with roles in cell cycle changes, apoptosis and inflammatory responses. This was in keeping with global DNA hypomethylation affecting genes involved in the same processes after chronic treatment. At the phenotypic level, we observed a dose-dependent decrease in NOK cell viability, induction of DNA damage, cell cycle changes and increased apoptosis, with the most pronounced effects observed under arsenic and SLT co-exposure conditions. Live-cell imaging experiments indicated that the DNA damage likely resulted from induction of apoptosis, an observation validated by a lack of exome-wide mutagenesis in response to chronic exposure to arsenic and/or smokeless tobacco. In sum, our integrative omics study provides novel insights into the acute and chronic responses to arsenic and smokeless tobacco (co-)exposure, with both types of responses converging on several key mechanisms associated with cancer hallmark processes. The resulting rich catalogue of molecular programs in oral cells regulated by arsenic and smokeless tobacco (co-)exposure may provide bases for future development of biomarkers for use in molecular cancer epidemiology studies of exposed populations at risk.
Collapse
Affiliation(s)
- Samrat Das
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Shefali Thakur
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Vincent Cahais
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - François Virard
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
- University Claude Bernard Lyon 1, INSERM U1052–CNRS UMR5286, Cancer Research Center, Centre Léon Bérard, Lyon, France
- University of Lyon, Faculty of Odontology, Hospices Civils de Lyon, Lyon, France
| | - Liesel Claeys
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Claire Renard
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Marie-Pierre Cros
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Stéphane Keïta
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Assunta Venuti
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Cécilia Sirand
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Akram Ghantous
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Michael Korenjak
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Jiri Zavadil
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
31
|
Chen J, Han H, Li L, Chen Z, Liu X, Li T, Wang X, Wang Q, Zhang R, Feng D, Yu L, Li X, Wang L, Li B, Li J. Prediction of cancer cell line-specific synergistic drug combinations based on multi-omics data. PeerJ 2025; 13:e19078. [PMID: 40028209 PMCID: PMC11869890 DOI: 10.7717/peerj.19078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Compared to single-drug therapy, combination therapy involves the use of two or more drugs to reduce drug dosage, decrease drug toxicity, and improve treatment efficacy. We developed an extreme gradient boosting (XGBoost)-based drug-drug cell line prediction model (XDDC) to predict synergistic drug combinations. XDDC was based on XGBoost and used one of the largest drug combination datasets, NCI-ALMANAC. In XDDC, drug chemical structures, adverse drug reactions, and target information were selected as drug features; gene expression, methylation, mutations, copy number variations, and RNA interference data were used as cell line features; and pathway information was incorporated to link drug features and cell line features. XDDC improved the interpretability of drug combination features and outperformed other machine learning methods. It achieved an area under the curve (AUC) of 0.966 ± 0.002 and an AUPR of 0.957 ± 0.002 when cross-validated on NCI-ALMANAC data. Different types of omics data were evaluated and compared in the model. Literature and experimental verification confirmed some of our predictions. XDDC could help medical professionals to rapidly screen synergistic drug combinations against specific cancer cell lines.
Collapse
Affiliation(s)
- Jiaqi Chen
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Huirui Han
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Lingxu Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Zhengxin Chen
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xinying Liu
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Tianyi Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xuefeng Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Qibin Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Ruijie Zhang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Dehua Feng
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Lei Yu
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Xia Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Limei Wang
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Bing Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| | - Jin Li
- College of Biomedical Information and Engineering, Kidney Disease Research Institute at the Second Affiliated Hospital, Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
32
|
Tshiyoyo KS, Rabbad A, Yusuf AA, Malgas S. Combination of citrus peel-derived essential oils with acarbose to inhibit amylolytic enzymes - A potential type II diabetes treatment approach. Int J Biol Macromol 2025; 306:141504. [PMID: 40015399 DOI: 10.1016/j.ijbiomac.2025.141504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Type 2 diabetes (T2D) can be managed by inhibiting amylolytic enzymes, α-amylase and α-glucosidase, reducing the impact of dietary carbohydrates on blood glucose elevation. Acarbose, a current α-glucose inhibitor (AGI), has excessive α-amylase inhibition, resulting in side effects associated with large amounts of undigested starch being fermented in the colon. This study evaluated the AGI efficacy of citrus peel-derived essential oils, where they were first tested in silico against the target amylolytic enzymes, and then their AGI activity was tested in vitro. The synergistic effects of the essential oils with acarbose against amylolytic enzymes were also determined. In silico and in vitro data of the efficacy of the essential oils as AGIs correlated positively; lower Ki values correlated with more negative binding affinity. Furthermore, molecular dynamic simulations of the most potent compounds were evaluated and indicated relative flexibility and stability induced upon ligand interactions with the protein. The standard AGI drug, acarbose, had the lowest Ki (0.10 ± 0.01 mg/mL) and more negative binding affinity (-7.5 kcal/mol) than the essential oils for α-glucosidase; however, the essential oils only showed potent inhibition against α-glucosidase, with the most potent essential oils being valencene (Ki = 0.33 ± 0.04 mg/mL), carveol (Ki = 0.53 ± 0.02 mg/mL) and geraniol (Ki = 0.56 ± 0.02 mg/mL). The essential oils and acarbose displayed competitive inhibition of α-glucosidase. Furthermore, a combination of acarbose with carveol or geraniol at a ratio of 12.5 μg/mL: 2 mg/mL exhibited antagonistic (CI > 10) and synergistic (CI < 0.7) effects on α-amylase and α-glucosidase inhibition, respectively. Carveol or geraniol can be considered as potentially therapeutic in managing T2D, as it may display lowered AGI-associated side effects.
Collapse
Affiliation(s)
- Kadima Samuel Tshiyoyo
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Ali Rabbad
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Abdullahi Ahmed Yusuf
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| | - Samkelo Malgas
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
33
|
Ziener J, Henao-Restrepo JA, Leonhardi J, Sturm MJ, Becker S, Morales-Prieto DM, Milde T, Beck JF, Sonnemann J. Combined inhibition of ribonucleotide reductase and WEE1 induces synergistic anticancer activity in Ewing's sarcoma cells. BMC Cancer 2025; 25:277. [PMID: 39962391 PMCID: PMC11831844 DOI: 10.1186/s12885-025-13691-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Ewing's sarcoma is a childhood bone and soft tissue cancer with poor prognosis. Treatment outcomes for Ewing's sarcoma patients have improved only modestly over the past decades, making the development of new treatment strategies paramount. In this study, the combined targeting of ribonucleotide reductase (RNR) and WEE1 was explored for its effectiveness against Ewing's sarcoma cells. METHODS The RNR inhibitor triapine and the WEE1 inhibitors adavosertib and ZN-c3 were tested in p53 wild-type and p53 mutant Ewing's sarcoma cells. The combination of adavosertib with the PARP inhibitors olaparib and veliparib was tested for comparison. Combinatorial effects were determined by flow cytometric analyses of cell death, loss of mitochondrial membrane potential and DNA fragmentation as well as by caspase 3/7 activity assay, immunoblotting and real-time RT-PCR. The drug interactions were assessed using combination index analysis. RESULTS RNR and WEE1 inhibitors were weakly to moderately effective on their own, but highly effective in combination. The combination treatments were similarly effective in p53 wild-type and p53 mutant cells. They synergistically induced cell death and cooperated to elicit mitochondrial membrane potential decay, to activate caspase 3/7 and to trigger DNA fragmentation, evidencing the induction of the apoptotic cell death cascade. They also cooperated to boost CHK1 phosphorylation, indicating augmented replication stress after combination treatment. In comparison, the combination of adavosertib with PARP inhibitors produced weaker synergistic effects. CONCLUSION Our findings show that combined inhibition of RNR and WEE1 was effective against Ewing's sarcoma in vitro. They thus provide a rationale for the evaluation of the potential of combined targeting of RNR and WEE1 in Ewing's sarcoma in vivo.
Collapse
Affiliation(s)
- Judy Ziener
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Johanna Leonhardi
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Max-Johann Sturm
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Sabine Becker
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
| | | | - Till Milde
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - James F Beck
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jürgen Sonnemann
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany.
- Klinik für Kinder- und Jugendmedizin, Friedrich-Schiller-Universität Jena, Am Klinikum 1, D-07747, Jena, Germany.
| |
Collapse
|
34
|
Erdogan MK, Ozer G. Synergistic Anticancer Effects of Bleomycin and Hesperidin Combination on A549 Non-Small Cell Lung Cancer Cells: Antiproliferative, Apoptotic, Anti-Angiogenic, and Autophagic Insights. Pharmaceuticals (Basel) 2025; 18:254. [PMID: 40006067 PMCID: PMC11859711 DOI: 10.3390/ph18020254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Background: This study investigated the combined effects of hesperidin (Hesp), a natural flavonoid, with bleomycin (BL), a commonly used chemotherapy agent, on A549 human lung cancer cells. Methods: Key parameters assessed included cell viability, colony formation, and cell migration, alongside the expression of apoptotic and autophagic markers (p53, p21, Bax, cleaved PARP, and Beclin-1), VEGF levels, and caspase-3 activity. Results: The findings revealed that the Hesp + BL combination significantly amplified antiproliferative, apoptotic, anti-angiogenic, and autophagic effects compared to either treatment alone. The combination therapy effectively inhibited colony formation and cell migration while markedly reducing VEGF levels, indicating strong anti-angiogenic properties. Apoptotic markers such as p53, p21, Bax, and cPARP were significantly upregulated, with caspase-3 activity confirming robust apoptosis induction. Furthermore, autophagy was notably enhanced, as reflected by increased Beclin-1 expression. Conclusions: Synergistic interactions between Hesp and BL, validated through combination index analysis, underscore the therapeutic potential of this combination. These findings underscore the therapeutic potential of the Hesp + BL combination as a promising strategy for lung cancer treatment, meriting further exploration in diverse lung cancer cell lines to validate and expand its applicability in developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, 12000 Bingol, Türkiye
| | - Guleser Ozer
- Department of Biology, Science Institute, Bingol University, 12000 Bingol, Türkiye
| |
Collapse
|
35
|
Hernando G, Turani O, Rodriguez Araujo N, Pulido Carrasquero A, Bouzat C. Unraveling anthelmintic targets and mechanisms of action of trans-cinnamaldehyde from cinnamon essential oil. Sci Rep 2025; 15:5422. [PMID: 39948358 PMCID: PMC11825704 DOI: 10.1038/s41598-025-89883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/10/2025] [Indexed: 02/16/2025] Open
Abstract
Parasitic nematodes pose a significant global socio-economic threat and contribute to neglected diseases. Current infection control relies on drug therapy, but increasing anthelmintic resistance highlights the urgent need for novel treatments. In this study, we investigate the molecular targets and mechanisms of action of trans-cinnamaldehyde (TCA), a principal component of Cinnamon Essential Oil (Cinnamomum verum EO), using Caenorhabditis elegans as a model organism. Our research offers new insights into the anthelmintic effects of TCA by identifying its specific interactions with key Cys-loop receptors and detailing its inhibitory mechanisms. The anthelmintic activity of C. verum EO and TCA manifests as rapid alterations in locomotor activity and inhibition of egg hatching. TCA screening of mutant worms lacking Cys-loop receptors reveal multiple receptor targets, including the levamisole-sensitive nicotinic ACh receptor (L-AChR), GABA-activated chloride channel (UNC-49) and glutamate-activated chloride channel. The mechanism behind the egg hatching inhibition by TCA remains unclear, as none of the mutants studied were found to be resistant to TCA. Furthermore, TCA increases the paralyzing effects of the anthelmintics levamisole and monepantel in a synergistic manner, offering a route for more effective polytherapy strategies. Electrophysiological studies on C. elegans Cys-loop receptors, in both native and heterologous systems, were used to elucidate the molecular mechanisms of TCA-induced paralysis. TCA reduces ACh- and GABA-elicited macroscopic currents and decreases single-channel activity and open durations of native muscle L-AChR channels, indicating an inhibitory action. Thus, by acting through a different mechanism to that of classical anthelmintics, TCA may be beneficial to counteract resistance in combined anthelmintic therapies. Our findings underscore the potential of the multitarget compound TCA as a valuable tool in integrated pharmacological strategies.
Collapse
Affiliation(s)
- Guillermina Hernando
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, 8000, Argentina.
| | - Ornella Turani
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, 8000, Argentina
| | - Noelia Rodriguez Araujo
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, 8000, Argentina
| | - Alcibeth Pulido Carrasquero
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, 8000, Argentina
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, Bahía Blanca, 8000, Argentina
| |
Collapse
|
36
|
Koh LWH, Pang QY, Novera W, Lim SW, Chong YK, Liu J, Ang SYL, Loh RWY, Shao H, Ching J, Wang Y, Yip S, Tan P, Li S, Low DCY, Phelan A, Rosser G, Tan NS, Tang C, Ang BT. EZH2 functional dichotomy in reactive oxygen species-stratified glioblastoma. Neuro Oncol 2025; 27:398-414. [PMID: 39373211 PMCID: PMC11812038 DOI: 10.1093/neuonc/noae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Enhancer of zeste homolog 2 (EZH2), well known for its canonical methyltransferase activity in transcriptional repression in many cancers including glioblastoma (GBM), has an understudied noncanonical function critical for sustained tumor growth. Recent GBM consortial efforts reveal complex molecular heterogeneity for which therapeutic vulnerabilities correlated with subtype stratification remain relatively unexplored. Current enzymatic EZH2 inhibitors (EZH2inh) targeting its canonical su(var)3-9, enhancer-of-zeste and trithorax domain show limited efficacy and lack durable response, suggesting that underlying differences in the noncanonical pathway may yield new knowledge. Here, we unveiled dual roles of the EZH2 CXC domain in therapeutically distinct, reactive oxygen species (ROS)-stratified tumors. METHODS We analyzed differentially expressed genes between ROS classes by examining cis-regulatory elements as well as clustering of activities and pathways to identify EZH2 as the key mediator in ROS-stratified cohorts. Pull-down assays and CRISPR knockout of EZH2 domains were used to dissect the distinct functions of EZH2 in ROS-stratified GBM cells. The efficacy of NF-κB-inducing kinase inhibitor (NIKinh) and standard-of-care temozolomide was evaluated using orthotopic patient-derived GBM xenografts. RESULTS In ROS(+) tumors, CXC-mediated co-interaction with RelB drives constitutive activation of noncanonical NF-κB2 signaling, sustaining the ROS(+) chemoresistant phenotype. In contrast, in ROS(-) subtypes, Polycomb Repressive Complex 2 methyltransferase activity represses canonical NF-κB. Addressing the lack of EZH2inh targeting its nonmethyltransferase roles, we utilized a brain-penetrant NIKinh that disrupts EZH2-RelB binding, consequently prolonging survival in orthotopic ROS(+)-implanted mice. CONCLUSIONS Our findings highlight the functional dichotomy of the EZH2 CXC domain in governing ROS-stratified therapeutic resistance, thereby advocating for the development of therapeutic approaches targeting its noncanonical activities and underscoring the significance of patient stratification methodologies.
Collapse
Affiliation(s)
- Lynnette Wei Hsien Koh
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Qing You Pang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Wisna Novera
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - See Wee Lim
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Yuk Kien Chong
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
| | - Jinyue Liu
- Laboratory of Single-Cell Spatial Neuromics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Samantha Ya Lyn Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- SingHealth Duke-NUS Neuroscience Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore, Singapore
| | | | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianhong Ching
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- KK Research Centre, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Yulan Wang
- Singapore Phenome Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shang Li
- Cancer and Stem Cell Biology Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Chyi Yeu Low
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- SingHealth Duke-NUS Neuroscience Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore, Singapore
| | | | | | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Carol Tang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Enabling Village, SG Enable, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Beng Ti Ang
- Neuro-Oncology Research Laboratory, Department of Research, National Neuroscience Institute, Singapore, Singapore
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
- SingHealth Duke-NUS Neuroscience Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore, Singapore
| |
Collapse
|
37
|
Zeng Y, Yang J, Li Y. Compartment Model and Neural Network-Based Analysis of Combination Medication Ratios. Pharmaceutics 2025; 17:228. [PMID: 40006595 PMCID: PMC11859217 DOI: 10.3390/pharmaceutics17020228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Combination medication strategies often involve complex interactions, making determining the appropriate pharmacodynamic component ratios challenging. Methods: This study established a time-dose relationship model through the compartment model, deriving the in vivo drug quantity ratios corresponding to the blood concentrations of the pharmacodynamic components. A neural network was then employed to establish a dose-effect relationship model between the blood concentrations of the pharmacodynamic components and the overall body response. Utilizing the feedback adjustment mechanism of neural networks continuously adjusts the network to achieve the desired drug efficacy, thereby deriving the corresponding dose ratio of the pharmacodynamic components. Empirical studies were conducted on combining Cynanchum otophyllum saponins M1 and M2 with phenobarbital for epilepsy treatment, as well as the anti-ischemic stroke activity of the prototype and metabolites of Erigeron breviscapus. Results: After adjusting the efficacy, the model recalculated the new ratio proportions for each combination, validated by the reduced Combination Index (CI). Conclusions: This model provides a new approach to combination medication strategies.
Collapse
Affiliation(s)
| | | | - Yong Li
- Faculty of Information Engineering and Automation, Kunming University of Science and Technology, Kunming 650500, China; (Y.Z.); (J.Y.)
| |
Collapse
|
38
|
Gutheil WG. Checkerboard synergistic data analysis using a Hill function-based approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636765. [PMID: 39975223 PMCID: PMC11839125 DOI: 10.1101/2025.02.05.636765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
This study presents a checkerboard data analysis approach using a Hill function (y = 1/(1+(x/K)n) to fit each column and row of checkerboard assay data. Column fits give a K (MIC) value in units of row concentration for each column antibiotic concentration (MIC_row vs [Col]), and row fits give an MIC_col value for each row antibiotic (MIC_row vs [Row]). Since the corresponding row and column concentrations are themselves MICs, this provides two sets of MIC vs MIC data pairs which can be plotted together as an isobologram. These MIC_A vs MIC_B values can be subjected to a second round of Hill function fitting, separately in x-y and y-x directions. Finally, a fit based on overlapping Hill functions is developed that allows x-y and y-x dimension fits to be performed simultaneously. Formula for fractional inhibitory concentrations (FICIs) as a function of fit parameters, and other features of these curves, are derived. This analysis also provides "n" (steepness) values from column and row fits, which are themselves dependent on the other antibiotic concentration and can be exceptionally, as in the case of ceftobiprole alone (n>10). This synergistic checkerboard analysis approach is implemented in MATLAB, which performs the fits and provides statistics variable values and alternative models significance.
Collapse
Affiliation(s)
- William G. Gutheil
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| |
Collapse
|
39
|
Erdogan MK, Usca AB. Gallic Acid Enhances Olaparib-Induced Cell Death and Attenuates Olaparib Resistance in Human Osteosarcoma U2OS Cell Line. Curr Issues Mol Biol 2025; 47:104. [PMID: 39996825 PMCID: PMC11854715 DOI: 10.3390/cimb47020104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/28/2024] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cancer remains one of the most formidable diseases globally and continues to be a leading cause of mortality. While chemotherapeutic agents are crucial in cancer treatment, they often come with severe side effects. Furthermore, the development of acquired drug resistance poses a significant challenge in the ongoing battle against cancer. Combining these chemotherapeutic agents with plant-derived phenolic compounds offers a promising approach, potentially reducing side effects and counteracting drug resistance. Phytochemicals, the bioactive compounds found in plants, exhibit a range of health-promoting properties, including anticarcinogenic, antimutagenic, antiproliferative, antioxidant, antimicrobial, neuroprotective, and cardioprotective effects. Their ability to enhance treatment, coupled with their non-toxic, multi-targeted nature and synergistic potential when used alongside conventional drugs, underscores the growing importance of natural therapeutics. In this study, we investigated the anticancer effects of olaparib (OL), a small-molecule PARP inhibitor that has shown promising results in both preclinical and clinical trials, and gallic acid (GA), a phenolic compound, in olaparib-resistant human osteosarcoma U2OS cells (U2OS-PIR). Both parental U2OS and U2OS-PIR cell lines were treated with increasing concentrations of olaparib and gallic acid, and their cytotoxic effects were assessed using the WST-1 cell viability assay. The synergistic potential of OL and GA, based on their determined IC50 values, was further explored in combination treatment. A colony survival assay revealed the combination's ability to significantly reduce the colony-forming capacity of cancer cells. Additionally, the apoptotic effects of OL and GA, both individually and in combination, were examined in U2OS-PIR cells using acridine orange/ethidium bromide dual staining. The anti-angiogenic properties were assessed through a VEGF ELISA, while the expression of proteins involved in DNA damage and apoptotic signaling pathways was analyzed via Western blot. The results of this study demonstrate that gallic acid effectively suppresses cell viability and colony formation, particularly when used in combination therapy to combat OL resistance. Additionally, GA inhibits angiogenesis and induces DNA damage and apoptosis by modulating key apoptosis-related proteins, including cPARP, Bcl-2, and Bax. These findings highlight gallic acid as a potential compound for enhancing therapeutic efficacy in overcoming acquired drug resistance.
Collapse
Affiliation(s)
- Mehmet Kadir Erdogan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Bingol University, Bingol 12000, Türkiye
| | - Ayse Busra Usca
- Department of Biology, Science Institute, Bingol University, Bingol 12000, Türkiye
| |
Collapse
|
40
|
Díaz-García A, Garrido Á, Ruiz-Fuentes JL, Hermosilla T, Varela D. Rhopalurus junceus scorpion venom induces G2/M cell cycle arrest and apoptotic cell death in human non-small lung cancer cell lines. J Venom Anim Toxins Incl Trop Dis 2025; 31:e20240035. [PMID: 39906356 PMCID: PMC11792888 DOI: 10.1590/1678-9199-jvatitd-2024-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 12/18/2024] [Indexed: 02/06/2025] Open
Abstract
Background Non-small cell lung cancers (NSCLC) represent the primary cause of cancer-related deaths worldwide. Rhopalurus junceus venom has been shown to exert cytotoxic effects against a panel of epithelial cancer cells in vitro and suggested that NSCLC was the subtype most susceptible to the treatment. Methods This study evaluated the effect of Rhopalurus junceus scorpion venom on cell viability, in non-cancerous (MRC-5, lung; CHO-K1, ovary) and NSCLC (A549; NCI-H460) cell lines. The effects on cell cycle, apoptosis, and cell signaling-related proteins were determined by flow cytometry and WB. Protein fractions responsible for the observed effect were identified using HPLC. Results Scorpion venom was more effective against NSCLC than non-cancerous cells. Emax values were 20.0 ± 5.8% and 22.47 ± 6.02% in A549 and NCI-H460 cancer cells, respectively, as compared to 50 ± 8.1% in MRC-5 and 54.99 ± 7.39% in CHO-K1 cells. It arrested NSCLC cells in the G2/M phase, while non-cancerous cells were arrested in the S (MRC-5) or G0/G1 (CHO-K1) phases. No changes were observed in the Bax/Bcl-2 or the cleaved-caspase 3/Total caspase 3 ratios in cells treated with venom. Likewise, the scorpion venom treatment did not affect p-ERK, p-AKT, or p-38MAPK protein levels. In contrast, scorpion venom treatment increased the cytosolic apoptosis-inducing factor (AIF) in A549 cells, indicating caspase-independent apoptosis. Additionally, combined etoposide/venom exposure provoked G2/M arrest and apoptosis in NSCLC more strongly than either substance alone. Furthermore, upon crude venom fractioning through RP-HPLC, we found two soluble fractions with high cytotoxic effects. Conclusion The present study concludes that a specific fraction of Rhopalurus junceus venom reduces cell viability of NSCLC cells. The AIF protein plays a key role in mediating caspase-independent apoptotic cell death. These findings suggest that Rhopalurus junceus venom enhances the anticancer effect of etoposide in vitro by causing cell cycle arrest and caspase-independent apoptosis.
Collapse
Affiliation(s)
- Alexis Díaz-García
- LifEscozul Chile, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),
University of Chile, Santiago, Chile
| | - Ángel Garrido
- LifEscozul Chile, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),
University of Chile, Santiago, Chile
| | - Jenny Laura Ruiz-Fuentes
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),
University of Chile, Santiago, Chile
- Physiology and Biophysics Program, Institute of Biomedical Sciences,
Faculty of Medicine, University of Chile, Santiago, Chile
| | - Tamara Hermosilla
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),
University of Chile, Santiago, Chile
- Physiology and Biophysics Program, Institute of Biomedical Sciences,
Faculty of Medicine, University of Chile, Santiago, Chile
| | - Diego Varela
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD),
University of Chile, Santiago, Chile
- Physiology and Biophysics Program, Institute of Biomedical Sciences,
Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
41
|
Rodriguez‐Galvez R, Tvarijonaviciute A, Peres‐Rubio C, Lopez‐Jornet P. Efficacy of Combined vs. Monotherapy in Oral Lichen Planus: A Randomized Clinical Trial. Oral Dis 2025; 31:522-531. [PMID: 39435599 PMCID: PMC11976123 DOI: 10.1111/odi.15151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVES The aim of this study was to compare the topical therapeutic efficacy of triamcinolone acetonide (TA) (0.2%) plus hyaluronic acid (HA) (0.1%) versus monotherapy in patients with symptomatic oral lichen planus (OLP), as well as to investigate the oxidative stress of saliva under the different treatments. MATERIALS AND METHODS Sixty OLP patients were included in a randomized, double-blind, singlecenter study with a treatment duration of 28 days and 3-month follow-up period. Participants were randomized into three groups: Group I (TA + HA), Group II (TA), and Group III (HA). Treatment efficacy was assessed by means of Thongprasom scale, Oral Health Impact Profile-14 (OHIP-14), and visual analog scale (VAS). In addition, biochemical analyses were performed in order to determine the level of antioxidant biomarkers in saliva, including superoxide dismutase (SOD), glutathione (GSH), and total antioxidant capacity (TAC). RESULTS All treatments seem to exhibit a significant effect in accordance with Thongprasom scale (p < 0.001), VAS reduction (p < 0.001), and OHIP14 (p < 0.05), which maintains over time. No significant changes in salivary oxidative stress in any of the three groups occurred. CONCLUSIONS The results exhibited a significant improvement in the treated patients in all three groups. There were no significant changes in salivary stress biomarkers under treatment condition.
Collapse
Affiliation(s)
| | - Asta Tvarijonaviciute
- Interdisciplinary Laboratory of Clinical Analysis (Interlab‐UMU), Veterinary School, Regional Campus of International Excellence Mare NostrumUniversity of MurciaMurciaSpain
| | - Camila Peres‐Rubio
- Interdisciplinary Laboratory of Clinical Analysis (Interlab‐UMU), Veterinary School, Regional Campus of International Excellence Mare NostrumUniversity of MurciaMurciaSpain
| | - Pia Lopez‐Jornet
- Faculty of Medicine and OdontologyHospital Morales MeseguerMurciaSpain
| |
Collapse
|
42
|
Tang JL, Moonshi SS, Wu Y, Cowin G, Vazquez- Prada KX, Tran HD, Bulmer AC, Ta HT. A methotrexate labelled dual metal oxide nanocomposite for long-lasting anti-cancer theranostics. Mater Today Bio 2025; 30:101377. [PMID: 39742148 PMCID: PMC11683249 DOI: 10.1016/j.mtbio.2024.101377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
We explored the feasibility of a self-assembled chitosan nanocomposite incorporating cerium oxide/nanoceria and superparamagnetic iron oxide nanoparticles (Chit-IOCO NPs), conjugated with methotrexate (MTX) and Cy5 dye, as an integrated cancer theranostic nanosystem (Chit-IOCO-MTX-Cy5). In this system, nanoceria serves as an anti-cancer agent, while the superparamagnetic iron oxide nanoparticles function as a negative contrast agent for MR imaging. This dual metal oxide nanocomposite is conjugated with MTX which is a structural analogue of folate, serving both as a targeting mechanism for folate receptors on cancer cells and as a chemotherapeutic drug. Chit-IOCO-MTX-Cy5 exhibited exceptional negative contrast in T2 and T2∗-weighted MRI, achieving a high relaxivity of 409.5 mM⁻1 s⁻1 which is superior to clinically approved agents. The nanocomposite demonstrated both pro-oxidative and antioxidative properties, significantly increasing reactive oxygen species (ROS) production in U87MG cells (1.4-fold change), which triggered apoptosis in these cancer cells. Simultaneously, it exhibited ROS scavenging activity in non-malignant endothelial cells (0.8-fold change). Intravenous infusion of Chit-IOCO-MTX-Cy5 (5 mg/kg MTX) led to significant tumor growth inhibition, indicating a synergistic enhancement of anti-cancer effects when combining MTX and nanoceria, compared to free MTX or nanoceria without MTX conjugation. Importantly, after treatment cessation, tumours in the nanocomposite group did not re-grow, while those in the free MTX group rapidly did. In vivo MR and fluorescence imaging revealed improved uptake and retention of Chit-IOCO-MTX-Cy5 in tumours compared to nanoceria without MTX. Notably, biosafety and biochemical analyses in mice showed no significant differences between the Chit-IOCO-MTX-Cy5 treatment group and control groups.
Collapse
Affiliation(s)
- Joyce L.Y. Tang
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Shehzahdi S. Moonshi
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Yuao Wu
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| | - Gary Cowin
- National Imaging Facility, Centre for Advanced Imaging, University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Karla X. Vazquez- Prada
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Huong D.N. Tran
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, Queensland, 4072, Australia
| | - Andrew C. Bulmer
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, 4215, Australia
| | - Hang Thu Ta
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, Queensland, 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, 4111, Australia
| |
Collapse
|
43
|
Hernando G, Bouzat C. Drug combination assays using Caenorhabditis elegans as a model system. J Pharmacol Toxicol Methods 2025; 131:107583. [PMID: 39855609 DOI: 10.1016/j.vascn.2025.107583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
The C. elegans drug combination assay evaluates the effects of drug combinations in the nematode Caenorhabditis elegans, serving as a valuable tool to assess the efficacy of pharmaceutical agents and natural compounds. Using C. elegans as a model organism, this method allows for the efficient screening of the combined effects of different drugs and evaluation of synergistic effects in drug combinations, which reduces the risk of developing drug resistance. Combination therapy, involving commercial drugs, new agents, or natural products, broadens treatment effectiveness by targeting multiple pathways, effectively managing complex diseases with minimized side effects. The method focuses on discovering effective drug combinations, such as anthelmintic drugs, streamlining early-stage drug discovery to save time and resources. Additionally, its versatility allows for application across most areas of pharmacology and toxicology, extending its usefulness beyond anthelmintic treatments. In the experiments, synchronized worms are exposed to different drug concentrations to evaluate behavioral changes, mostly alterations in worm locomotion. Concentration-response curves for changes in behavior are generated and EC50 or IC50 values determined for the individual drugs. To determine whether the effects of a drug combination are synergistic, additive, or antagonistic, at least three different concentration ratios must be tested. These combinations are then analyzed using specialized drug combination analysis software. This methodology ensures consistent and precise outcomes and evaluates drug impacts on worm behavior parameters crucial for effective pharmacological activity. In conclusion, the C. elegans drug combination assay provides critical insights for developing successful market formulations applicable across a wide range of pharmacological treatments. Its ability to efficiently screen for synergistic, additive, or antagonistic effects makes it a valuable tool for identifying effective therapeutic strategies, potentially reducing drug resistance and improving treatment outcomes in various medical and toxicological fields.
Collapse
Affiliation(s)
- Guillermina Hernando
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, 8000 Bahía Blanca, Argentina.
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS)-CONICET, 8000 Bahía Blanca, Argentina
| |
Collapse
|
44
|
Irum I, Khan F, Sufyan M, Benish Ali SH, Rehman S. Developing multifaceted drug synergistic therapeutic strategy against neurological disorders. Comput Biol Med 2025; 185:109495. [PMID: 39693689 DOI: 10.1016/j.compbiomed.2024.109495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Drug synergism can alter the ultimate biological effects and bioavailability of phytoconstituents. Acetylcholinesterase (AChE) inhibitors as symptomatic drugs are potent therapeutic regimen for neurodegenerative diseases. In this context, this study characterized the synergistic antioxidant, anti-inflammatory and anti-AChE effects of the selected phytochemicals including standard drugs followed by enzyme kinetics, structure-based ligands screening and molecular dynamics simulation study. The synergistic interactions were evaluated through Isoradiation and Synergy finder 3.0 methods. The combinations of Quercetin (QCT), Folic acid (FA), and Swertiamarin (SWT) with specific reference drugs were studied. The combinations of SWT + GA (Gallic acid) and FA + GA at 1:1 (γ:0.10 & 0.08, respectively) showed the significant synergistic antioxidant effect via ABTS assay. Further, in combination, QCT + SWT showed the maximum synergistic effect (γ: 0.02-0.13) in anti-inflammatory assay. Moreover, the combinations QCT, FA, and SWT with reference drug, Donepezil (DP), illustrated potent synergistic activity as anti-AChE in 1:1 proportion (γ: 0.18). The interaction pattern of phytochemicals significantly exhibited synergism (γ < 1) depicting their optimum activity in combinations compared to individual components. Enzyme kinetics evaluation showed the competitive binding of SWT with AChE as of donepezil. All the parameters of ADMET study proposed the QCT and SWT as acceptable oral drug molecules. Computational docking study revealed that QCT and SWT with lowest RMSD (1.096, 2.104) and lowest docking score (-9.831, -7.435 kcal/mol) showed maximum binding efficacy. Furthermore, molecular simulation study depicted the stability of protein-ligand complexes. These findings provide novel insight in the development of dietary treatment based on their synergistic effects for neurological disorders as optimum alternative therapeutic agents.
Collapse
Affiliation(s)
- Izza Irum
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan
| | - Fariha Khan
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan
| | - Muhammad Sufyan
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Syeda Hafiza Benish Ali
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan
| | - Sidra Rehman
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan.
| |
Collapse
|
45
|
Banchi M, Cox MC, Bandini A, Orlandi P, Tacchi C, Stefanelli F, Chericoni S, Bocci G. Linifanib alone and in combination with metronomic chemotherapy is active on cutaneous T-cell lymphoma cells by targeting the AKT/mTOR signaling pathway. Invest New Drugs 2025; 43:135-146. [PMID: 39821757 DOI: 10.1007/s10637-024-01501-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
Cutaneous T-cell lymphomas (CTCLs) are a rare and heterogeneous subset of skin-localized, non-Hodgkin lymphomas. Our aim was to evaluate the in vitro antitumor activity of the multi-kinase inhibitor linifanib, either alone or in combination with metronomic vinorelbine (mVNR) or etoposide (mETO), on CTCL cells. In vitro proliferation assay and Luminex analysis showed that long-term, daily exposure of linifanib significantly inhibited the proliferation of the human CTCL cell line HH, in a concentration-dependent manner (IC50 = 48.4 ± 20.4 nM) and the phosphorylation of AKT/mTOR signaling pathway. The concomitant exposure of linifanib plus mVNR or mETO resulted in a strong synergism, with combination index values < 1. Linifanib significantly increased the VNR and ETO intracellular concentrations in HH cells, evaluated by UPLC-HRMS technology, and strongly reduced the ABCB1 and ABCG2 gene expression in HH. In conclusion, we reported a striking antitumor activity of daily, long-term linifanib and a clear synergistic effect when administered in combination with mCHEMO on CTCL cells, as a promising base for future clinical approaches in T-cell lymphomas.
Collapse
Affiliation(s)
- Marta Banchi
- Dipartimento Di Ricerca Traslazionale E Delle Nuove Tecnologie in Medicina E Chirurgia, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy.
| | - Maria Christina Cox
- Unità Di Ematologia, Fondazione Policlinico Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Arianna Bandini
- Dipartimento Di Ricerca Traslazionale E Delle Nuove Tecnologie in Medicina E Chirurgia, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
- Dipartimento Di Medicina Clinica E Sperimentale, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Paola Orlandi
- Dipartimento Di Medicina Clinica E Sperimentale, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Costanza Tacchi
- Dipartimento Di Ricerca Traslazionale E Delle Nuove Tecnologie in Medicina E Chirurgia, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
- Dipartimento Di Medicina Clinica E Sperimentale, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Fabio Stefanelli
- Dipartimento Di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Silvio Chericoni
- Dipartimento Di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Guido Bocci
- Dipartimento Di Ricerca Traslazionale E Delle Nuove Tecnologie in Medicina E Chirurgia, Università Di Pisa, Via Savi 10, 56126, Pisa, Italy
| |
Collapse
|
46
|
Sawada JI, Matsuno K, Ogo N, Asai A. Identification of antimitotic sulfonamides inhibiting chromosome congression. Biochem Pharmacol 2025; 232:116718. [PMID: 39701545 DOI: 10.1016/j.bcp.2024.116718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
The discovery of new small-molecule inhibitors is essential to enhancing our understanding of biological events at the molecular level and driving advancements in drug discovery. Mitotic inhibitors have played a crucial role in development of anticancer drugs. Beyond traditional microtubule inhibitors, various inhibitors targeting specific mitotic factors have been developed. This study aimed to develop novel mitotic inhibitors targeting chromosome alignment. We established a cell-based screening method using Cell Division Cycle Associated 5 (CDCA5) and kinesin-5 as markers, designed to efficiently detect mitotic phenotypes characterized by aberrant bipolar spindles with some misaligned chromosomes. Through this screening, we identified CAIS-1, an aryl sulfonamide with unique antimitotic properties. CAIS-1 exhibits dual functionality by inhibiting chromosome congression at low concentrations and spindle microtubule formation at high concentrations, causing a concentration-dependent mitotic arrest, followed by apoptotic cell death. Mechanistic studies revealed that CAIS-1 directly acts on tubulin at high concentrations, thereby inhibiting tubulin polymerization in vitro. In contrast, at low concentrations, CAIS-1 functions through a mechanism distinct from GSK923295, a conventional chromosome congression inhibitor targeting Centromere-associated protein-E (CENP-E), highlighting its unique mode of action. Moreover, CAIS-2, a structural analog of CAIS-1, selectively inhibits chromosome congression without significantly affecting spindle microtubules. This observation suggests that CAIS-1 and CAIS-2 function as antimitotic sulfonamides with distinct targets beyond tubulin, thus offering additional biological potential of sulfonamide compounds. Together, CAIS-1 and CAIS-2 represent promising tools for providing new molecular insights into kinetochore function during mitosis and for exploring new approaches in anticancer drug development.
Collapse
Affiliation(s)
- Jun-Ichi Sawada
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Kenji Matsuno
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
47
|
Kotze AC, Knowles AG. Antagonistic interactions between spinosad and macrocyclic lactones in combination against larvae of the sheep blowfly, Lucilia cuprina, in vitro. Vet Parasitol 2025; 334:110389. [PMID: 39764947 DOI: 10.1016/j.vetpar.2024.110389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 02/27/2025]
Abstract
Control of the sheep blowfly relies on insecticides, however resistance is currently impacting on their efficacy. The use of insecticides in combination (mixtures) is considered to be a useful strategy to delay resistance under some circumstances. The present study aimed to examine the combination of spinosad with macrocyclic lactones in order to determine if the two drug classes showed any interactions that would impact on the usefulness of a combination product for flystrike control. We used isobologram analysis to examine drug interactions in in vitro bioassays with the larval stages of the sheep blowfly. In initial single drug assays, ivermectin and abamectin showed similar EC50 against blowfly larvae, while moxidectin and spinosad were 20- and 30-fold less potent, respectively. Combinations of spinosad and each of the macrocyclic lactones showed a pattern of interactions dominated by antagonistic or additive relationships when combined at different ratios. Peaks in antagonism were associated with combinations at ratios equivalent to, or close to, the relative EC50s when used alone. The antagonism was strongest for moxidectin, with combination index values up to 1.82 at the EC50 and 3.2 at the EC90 at a spinosad: moxidectin ratio of 0.78:1. Maximum combination index values for ivermectin and abamectin at the EC90 were 1.61 and 1.42, at ratios of 27:1 and 31:1, respectively, indicating a significant degree of antagonism for both combinations. We suggest that the observed antagonism may be due to interactions at common ion channel receptors, although this requires confirmation. The study highlights the need to examine drug interactions as a component of determining the suitability of specific drug combinations for parasite control.
Collapse
Affiliation(s)
- Andrew C Kotze
- CSIRO Agriculture and Food, Brisbane, QLD 4069, Australia; School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia.
| | | |
Collapse
|
48
|
Miyazaki S, Kitazawa M, Nakamura S, Koyama M, Yamamoto Y, Hondo N, Kataoka M, Tanaka H, Takeoka M, Komatsu D, Soejima Y. Targeting KRAS-mutant pancreatic cancer through simultaneous inhibition of KRAS, MEK, and JAK2. Mol Oncol 2025; 19:377-390. [PMID: 39400496 PMCID: PMC11793007 DOI: 10.1002/1878-0261.13751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 08/02/2024] [Accepted: 10/03/2024] [Indexed: 10/15/2024] Open
Abstract
The Kirsten rat sarcoma (KRAS) oncogene was considered "undruggable" until the development of sotorasib, a KRASG12C selective inhibitor that shows favorable effects against lung cancers. MRTX1133, a novel KRASG12D inhibitor, has shown promising results in basic research, although its effects against pancreatic cancer are limited when used alone. Therefore, there is an urgent need to identify effective drugs that can be used in combination with KRAS inhibitors. In this study, we found that administration of the KRAS inhibitors sotorasib or MRTX1133 upregulated STAT3 phosphorylation and reactivated ERK through a feedback reaction. The addition of the MEK inhibitor trametinib and the JAK2 inhibitor fedratinib successfully reversed this effect and resulted in significant growth inhibition in vitro and in vivo. Analyses of sotorasib- and MRTX1133-resistant cells showed that trametinib plus fedratinib reversed the resistance to sotorasib or MRTX1133. These findings suggest that the JAK2-mediated pathway and reactivation of the MAPK pathway may play key roles in resistance to KRAS inhibitors in pancreatic cancers. Accordingly, simultaneous inhibition of KRAS, MEK, and JAK2 could be an innovative therapeutic strategy against KRAS-mutant pancreatic cancer.
Collapse
Affiliation(s)
- Satoru Miyazaki
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Masato Kitazawa
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Satoshi Nakamura
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Makoto Koyama
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Yuta Yamamoto
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Nao Hondo
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Masahiro Kataoka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Hirokazu Tanaka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | - Michiko Takeoka
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| | | | - Yuji Soejima
- Division of Gastroenterological, Hepato‐Biliary‐Pancreatic, Transplantation and Pediatric Surgery, Department of SurgeryShinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
49
|
Rani S, Gupta S, Tejavath KK, Gupta U. Effect of combination of polyphenols, polysaccharide, and sodium selenite on bortezomib anti-cancer action. Int J Biol Macromol 2025; 289:138809. [PMID: 39694374 DOI: 10.1016/j.ijbiomac.2024.138809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/22/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Combinatorial drug delivery has shown promising results over single drug for cancer therapy. Here, we aimed to explore combination of proteasome inhibitor; bortezomib (BTZ) with natural antioxidants (AOs); polyphenols like caffeic acid (CFA), resveratrol (RES), fucoidan (FD), and synthetic AO; sodium selenite (Na2SeO3) for cellular cytotoxicity in breast cancer cell lines; MCF-7 and MDA MB-231. The combination of RES + BTZ, FD + BTZ, and Na2SeO3 + BTZ showed synergism while CFA showed antagonism with BTZ. The EC50 values of different combinations were found to be significantly less than the individual AOs in ABTS and DPPH assay. Furthermore, the effect of combination of drugs on migratory properties of MCF-7 cells were evaluated by in-vitro wound healing assay, resulting in the reduction of such behavior. In support of this, RT-qPCR was performed to analyze differential gene expressions of apoptotic and Epithelial-Mesenchymal Transition (EMT) markers with and without treatment. In results, the combination of Na2SeO3 + BTZ reduced the expression of Bcl-XL and N-Cad causing cytotoxicity and suggested that the combination of Na2SeO3 + BTZ (IC50 = 1.40 ± 0.45 μM) could be a better option among other combinations for breast cancer therapy. Overall, the outcome indicates that the combination of BTZ with AOs may yield potential therapeutic benefit.
Collapse
Affiliation(s)
- Sarita Rani
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Shruti Gupta
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Kiran Kumar Tejavath
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India; Department of Biochemistry, All India Institute of Medical Sciences, BIBINAGAR, HYDERABAD METROPOLITAN REGION (HMR) TELANGANA 508126.
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
50
|
Medeiros MT, Campos DR, de Assis JD, de Oliveira GF, Ferreira TP, Teixeira de Jesus AC, Correia TR, Scott FB, Cid YP, Coumendouros K. Effects of combinations of the essential oils trans-anethole, thymol and carvacrol against larvae of the screwworm fly Cochliomyia hominivorax in vitro. Vet Parasitol 2025; 334:110398. [PMID: 39847831 DOI: 10.1016/j.vetpar.2025.110398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
This study investigated the combined effect of trans-anethole, carvacrol and thymol on third-instar larvae of C. hominivorax. For this experiment, third-stage larvae of C. hominivorax were removed from a laboratory colony to perform an in vitro bioassay. The three volatile compounds used in this study, trans-anethole, carvacrol and thymol. Different dilutions of these volatile compounds were performed with the diluent acetone (99.5 %) to determine the associative effect (synergism, addition or antagonism). Three concentrations below the estimated LC50 were prepared (10, 20 and 100 μg.cm-2). These compounds were impregnated in filter paper discs, which were dried and transferred to Petri dishes. Ten larvae were added to each dish and incubated in climate-controlled chambers at 28 ± 1 ºC and 75 ± 10 % relative humidity for 24 and 48 hours. Mortality was assessed based on the absence or change in larval movement, as observed under a stereomicroscope. After 24 hours, trans-anethole (A) caused mortality rates in the range of 23-33.5 %, carvacrol (C) of 9-59 % and thymol (T) of 27-81.5 %, in the three concentrations evaluated. The combination of trans-anethole with carvacrol resulted in mortality rates between 11 % and 50 %, demonstrating antagonistic action. The combination of trans-anethole with thymol caused mortality of 12.5-55.5 %, and the combination of carvacrol with thymol generated mortality of 75.5-99.5 %, indicating synergism. After 48 hours, (A) produced a mortality range of 29-49 %, (C) of 10.5-63 %, and (T) of 37.5-86 % in the three concentrations, maintaining the same relationship of antagonistic and synergistic activity. The combination of trans-anethole with carvacrol resulted in mortality rates between 16 % and 56.5 %, while the combination of trans-anethole with thymol caused mortality of 27.5-62.5 % and the combination of carvacrol with thymol presented mortality between 83 % and 99.5 %. Hence, there was an antagonistic effect when trans-anethole was associated with carvacrol and thymol and a dose-dependent synergistic effect when carvacrol was associated with thymol against third instar larvae of C. hominivorax.
Collapse
Affiliation(s)
- Monique Taveira Medeiros
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Diefrey Ribeiro Campos
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Jéssica Dávila de Assis
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Gabriela Ferreira de Oliveira
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Thais Paes Ferreira
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Ciências Farmacêuticas, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Anna Carolina Teixeira de Jesus
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil.
| | - Thais Ribeiro Correia
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Fabio Barbour Scott
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Yara Peluso Cid
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Ciências Farmacêuticas, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Katherina Coumendouros
- Laboratório de Quimioterapia Experimental em Parasitologia Veterinária (LQEPV), Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil; Departamento de Parasitologia Animal, Instituto de Medicina Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| |
Collapse
|