101
|
Skvortsov DA, Zhirkina IV, Ipatova DA, Vasilyeva LA, Ivanenkov YA, Rubtsova MP, Kartsev VG, Sergiev PV, Dontsova OA. Coculture-Based Screening Revealed Selective Cytostatic Effects of Pyrazol-Azepinoindoles. ChemMedChem 2025:e2500052. [PMID: 40159440 DOI: 10.1002/cmdc.202500052] [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: 01/20/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
This work focuses on the search for new small molecules for anticancer therapy using the fluorescent cells cocultivation test (FCCT). This method allows the control of the specificity of the action of compounds from the earliest stages of drug development. For the FCCT, labeled MCF7' breast cancer cells and noncancerous breast MCF10A cells are cocultured. Screening of 2025 compounds in the above system and previously developed coculture of A549 with VA13 yields 16 selectively cytotoxic molecules. The results are confirmed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for seven of these molecules. Few are known as potential antitumor agents: angelicin, coumarin, and colchicine derivatives. However, the structures of macrocycle 1, pyrazole-azepinoindole derivative 2, and complex heterocyclic derivative 3 are not described as anticancer compounds according to the PubChem and SciFinder databases. Structure-activity relationships are investigated for 2 and its derivatives. The indole with a caprolactam ring (tetrahydro-azepinoindolone core) together with the pyrazolyl at the third position is the key element of the pharmacophore. The optimized pyrazole-azepinoindole derivative 23 shows SI = 18 for HCT116 versus VA-13 on the expanded array of cell lines. Its effect is mainly mediated by the G1 arrest of the cell cycle.
Collapse
Affiliation(s)
- Dmitry A Skvortsov
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Irina V Zhirkina
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Daria A Ipatova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Lilya A Vasilyeva
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Yan A Ivanenkov
- P. Hertsen Moscow Oncology Research Institute, Moscow, 3125284, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russian Federation
| | - Maria P Rubtsova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | | | - Petr V Sergiev
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
- Center of Molecular and Cellular Biology, Moscow, 121205, Russian Federation
| | - Olga A Dontsova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
- Center of Molecular and Cellular Biology, Moscow, 121205, Russian Federation
| |
Collapse
|
102
|
Milutinović M, Ristanović F, Radenković N, Cvetković D, Radenković S, Stanković M, Nikodijević D. Modulation of the Main Resistance-Associated ABC Transporter's Expression by Plant Flavonol Isorhamnetin. Pharmaceuticals (Basel) 2025; 18:494. [PMID: 40283931 PMCID: PMC12029997 DOI: 10.3390/ph18040494] [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: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Multidrug resistance is one the leading problems in cancer treatment, where the overexpression of P-gp and other drug efflux pumps is regarded as the primary cause. With the intention to develop transporter inhibitors, natural products such as phenolics have shown great potential and diverse attention recently. Among these, isorhamnetin (ISO), an O-methylated flavonol, is predominantly found in the fruits and leaves of various plants. Thus, this study aimed to investigate the effects of ISO on the mRNA expression of membrane transporters P-gp, BCRP, MRP 1, 2, and 5, the protein expression of P-gp, as well as the GSTP1 and GSH content in DLD1 and HCT-116 colon cancer cells. Methods: The cytotoxic effect of isorhamnetin is assessed using an MTT test, while qPCR and immunocytochemistry methods were used to determine gene and protein expression levels. The concentration of reduced glutathione was determined using the colorimetric method. Results: Based on the results, ISO can modulate the expression of transporters responsible for the resistance development (all transporters on the transcriptional level were downregulated in DLD1 cells, while only MRP1 on HCT-116 cells, and reduced P-gp protein expression on both investigated cell lines). Increased glutathione content in treated cells and GSTP1 expression suggest metabolizing the ISO and potential ejection with GSH-dependent pumps. Conclusions: Thus, in future experiments, ISO as a natural medicinal compound could be used as a chemosensitizer to prevent or overcome membrane transporter-mediated drug resistance.
Collapse
Affiliation(s)
- Milena Milutinović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia (M.S.)
| | - Filip Ristanović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia (M.S.)
| | - Nikola Radenković
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia (M.S.)
| | - Danijela Cvetković
- Faculty of Medical Science, University of Kragujevac, P.O. Box 124, 34000 Kragujevac, Serbia
| | - Sandra Radenković
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Beograd, Serbia
| | - Milan Stanković
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia (M.S.)
| | - Danijela Nikodijević
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia (M.S.)
| |
Collapse
|
103
|
Logan JA, Sadhu S, Hazlewood C, Denton M, Burke SE, Simone-Soule CA, Black C, Ciaverelli C, Stulb J, Nourzadeh H, Vinogradskiy Y, Leader A, Dicker AP, Choi W, Simone NL. Bridging Gaps in Cancer Care: Utilizing Large Language Models for Accessible Dietary Recommendations. Nutrients 2025; 17:1176. [PMID: 40218934 PMCID: PMC11990115 DOI: 10.3390/nu17071176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Weight management is directly linked to cancer recurrence and survival, but unfortunately, nutritional oncology counseling is not typically covered by insurance, creating a disparity for patients without nutritional education and food access. Novel ways of imparting personalized nutrition advice are needed to address this issue. Large language models (LLMs) offer a promising path toward tailoring dietary advice to individual patients. This study aimed to assess the capacity of LLMs to offer personalized dietary advice to patients with breast cancer. Methods: Thirty-one prompt templates were designed to evaluate dietary recommendations generated by ChatGPT and Gemini with variations within eight categorical variables: cancer stage, comorbidity, location, culture, age, dietary guideline, budget, and store. Seven prompts were selected for four board-certified oncology dietitians to also respond to. Responses were evaluated based on nutritional content and qualitative observations. A quantitative comparison of the calories and macronutrients of the LLM- and dietitian-generated meal plans via the Acceptable Macronutrient Distribution Ranges and United States Department of Agriculture's estimated calorie needs was performed. Conclusions: The LLMs generated personalized grocery lists and meal plans adapting to location, culture, and budget but not age, disease stage, comorbidities, or dietary guidelines. Gemini provided more comprehensive responses, including visuals and specific prices. While the dietitian-generated diets offered more adherent total daily calorie contents to the United States Department of Agriculture's estimated calorie needs, ChatGPT and Gemini offered more adherent macronutrient ratios to the Acceptable Macronutrient Distribution Range. Overall, the meal plans were not significantly different between the LLMs and dietitians. LLMs can provide personalized dietary advice to cancer patients who may lack access to this care. Grocery lists and meal plans generated by LLMs are applicable to patients with variable food access, socioeconomic means, and cultural preferences and can be a tool to increase health equity.
Collapse
Affiliation(s)
- Julia A. Logan
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Sriya Sadhu
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Cameo Hazlewood
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Melissa Denton
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University Hospitals, Philadelphia, PA 19107, USA
| | - Sara E. Burke
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Christina A. Simone-Soule
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Caroline Black
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University Hospitals, Philadelphia, PA 19107, USA
| | - Corey Ciaverelli
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University Hospitals, Philadelphia, PA 19107, USA
| | - Jacqueline Stulb
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University Hospitals, Philadelphia, PA 19107, USA
| | - Hamidreza Nourzadeh
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Yevgeniy Vinogradskiy
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Amy Leader
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam P. Dicker
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Wookjin Choi
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| | - Nicole L. Simone
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (S.S.); (S.E.B.); (A.P.D.); (W.C.)
| |
Collapse
|
104
|
Andavar A, Bhagavathi VR, Cousin J, Parekh N, Razavi ZS, Tan B. Current Research in Drug-Free Cancer Therapies. Bioengineering (Basel) 2025; 12:341. [PMID: 40281701 PMCID: PMC12024433 DOI: 10.3390/bioengineering12040341] [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: 12/27/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer treatment has historically depended on conventional methods like chemotherapy, radiation, and surgery; however, these strategies frequently present considerable limitations, including toxicity, resistance, and negative impacts on healthy tissues. In addressing these challenges, drug-free cancer therapies have developed as viable alternatives, utilizing advanced physical and biological methods to specifically target tumor cells while reducing damage to normal tissues. This review examines several drug-free cancer treatment strategies, such as high-intensity focused energy beams, nanosecond pulsed electric fields, and photothermal therapy as well as the use of inorganic nanoparticles to promote selective apoptosis. We also investigate the significance of targeting the tumor microenvironment, precision medicine, and immunotherapy in the progression of personalized cancer therapies. Although these approaches demonstrate significant promise, challenges including scalability, safety, and regulatory obstacles must be resolved for clinical application. This paper presents an overview of current research in drug-free cancer therapies, emphasizing recent advancements, underlying scientific principles, and the steps required for clinical implementation.
Collapse
Affiliation(s)
- Akshaya Andavar
- Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | | | - Justine Cousin
- École Publique d’Ingénieurs de la Santé et du Numérique (EPISEN), Université Paris-Est Créteil (UPEC), 94000 Créteil, France;
| | - Nirvi Parekh
- Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai 400019, India;
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran 1416634793, Iran;
| | - Bo Tan
- Institute of Biomedical Engineering Science and Technology (iBEST), Faculty of Engineering and Architectural Science, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| |
Collapse
|
105
|
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
|
106
|
Eira A, Gonçalves MBS, Fongang YSF, Domingues C, Jarak I, Mascarenhas-Melo F, Figueiras A. Unlocking the Potential of Ganoderma lucidum (Curtis): Botanical Overview, Therapeutic Applications, and Nanotechnological Advances. Pharmaceutics 2025; 17:422. [PMID: 40284417 PMCID: PMC12030363 DOI: 10.3390/pharmaceutics17040422] [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: 01/20/2025] [Revised: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Background:Ganoderma lucidum (GL), commonly known as the "Lingzhi" or "Reishi" mushroom, has long been recognized for its potential health benefits and medicinal properties in traditional Chinese medicine. The unique potential combination of bioactive compounds present in GL, such as triterpenoids, polysaccharides, and peptides, has inspired interest in leveraging their therapeutic potential In recent years, the emerging field of nanotechnology has opened up new possibilities for using the remarkable properties of GL at the nanoscale. Objetive: The main objective of this review is to explore the unique potential of GL in traditional and innovative therapies, particularly in cancer treatment, and to assess how nanotechnology-based strategies can enhance its therapeutic applications.is to explore. Results: Nanotechnology-based strategies have been investigated for the efficient extraction and purification of bioactive compounds from GL. Additionally, nanocarriers and nanoformulations have been developed to protect these sensitive bioactive compounds from degradation, ensuring their stability during storage and transportation. The use of GL-based nanomaterials has shown promising results in several biomedical applications, namely due to their anticancer activity by targeting cancer cells, inducing apoptosis, and inhibiting tumor growth. Conclusions: The combination of GL and nanotechnology presents an exciting frontier in the development of novel therapeutic and biomedical applications. Nevertheless, further research and development in this interdisciplinary field are warranted to fully exploit the synergistic benefits offered by GL and nanotechnology. Future prospects include the development of robust clinical trials focused on GL nanotechnology-based cancer therapies to clarify mechanisms of actions and optimize formulations, ultimately leading to innovative solutions for human health and well-being.
Collapse
Affiliation(s)
- Ana Eira
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.E.); (M.B.S.G.); (C.D.); (I.J.)
| | - Maria Beatriz S. Gonçalves
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.E.); (M.B.S.G.); (C.D.); (I.J.)
| | | | - Cátia Domingues
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.E.); (M.B.S.G.); (C.D.); (I.J.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Group of Pharmaceutical Technology, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CI MAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ivana Jarak
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.E.); (M.B.S.G.); (C.D.); (I.J.)
| | - Filipa Mascarenhas-Melo
- Higher School of Health, Polytechnic Institute of Guarda, 6300-307 Guarda, Portugal
- BRIDGES—Biotechnology Research, Innovation and Design of Health Products, Polytechnic University of Guarda, 6300-559 Guarda, Portugal
| | - Ana Figueiras
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.E.); (M.B.S.G.); (C.D.); (I.J.)
- Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE), Group of Pharmaceutical Technology, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
107
|
Mehta D, Dua C, Chakraborty R, Yadav P, Dasgupta U, Bajaj A. Docetaxel-conjugated bile acid-derived nanomicelles can inhibit tumour progression with reduced toxicity. NANOSCALE ADVANCES 2025; 7:2003-2010. [PMID: 39967859 PMCID: PMC11832056 DOI: 10.1039/d4na00715h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Docetaxel (DTX) is a highly effective chemotherapy drug commonly employed in the management of multiple cancers, such as breast, lung, and prostate cancer. However, its clinical usage is significantly hampered by its limited solubility, which limits its bioavailability, and its considerable toxic effects like neutropenia, neuropathy, and hypersensitive reactions. These limitations necessitate the development of innovative formulations to boost the therapeutic index of DTX. In this study, we aimed to enhance the tolerability and reduce the toxic effects of DTX by developing a novel hybrid scaffold (PIP-LCA-DTX), where we conjugated DTX to piperidine-derived lithocholic acid. This hybrid scaffold integrates the beneficial properties of bile acid-based drug conjugates and cationic amphiphiles to form stable and effective drug delivery systems. Our research demonstrates that PIP-LCA-DTX exhibits similar anticancer properties to DTX when tested against murine colon cancer (CT26) and melanoma (B16-F10) cell lines, indicating that the hybrid retains the therapeutic efficacy of the original drug. Our findings revealed that PIP-LCA-DTX forms stable nanomicelles (DTX-NMs) with an average hydrodynamic diameter of <150 nm and provides a promising delivery system by enhancing the solubility and stability of DTX. DTX-NMs showed significantly better tolerability and enhanced therapeutic efficacy (survival) compared to DTX alone. This improved tolerability, combined with the maintained therapeutic efficacy of DTX-NMs against murine cancer models, suggests that this hybrid scaffold could offer a more viable and safer option for cancer treatment.
Collapse
Affiliation(s)
- Devashish Mehta
- Amity Institute of Biotechnology, Amity University Haryana Panchgaon, Manesar Gurgaon 122413 Haryana India
| | - Chhavi Dua
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Ruchira Chakraborty
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| | - Ujjaini Dasgupta
- Amity Institute of Biotechnology, Amity University Haryana Panchgaon, Manesar Gurgaon 122413 Haryana India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 Haryana India
| |
Collapse
|
108
|
Ojo OA, Grant S, Nwafor-Ezeh PI, Maduakolam-Aniobi TC, Akinborode TI, Ezenabor EH, Ojo AB. Ferroptosis as the new approach to cancer therapy. Cancer Treat Res Commun 2025; 43:100913. [PMID: 40187205 DOI: 10.1016/j.ctarc.2025.100913] [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: 11/26/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025]
Abstract
Cancer is characterized by unregulated cell proliferation, evasion of apoptosis, and a propensity for metastasis, making it a leading cause of morbidity and mortality globally. Major challenges in cancer treatment include drug resistance and tumor heterogeneity, which hinder the clinical efficacy of existing therapies. To enhance treatment outcomes, it is essential to integrate emerging biological insights and technological advancements with conventional therapeutic strategies. Recent research has identified various forms of cell death, which can be classified as either regulated or unregulated. Regulated cell death involves specific biochemical and signaling pathways, while unregulated cell death occurs passively and uncontrollably. Apoptosis, the most extensively studied form of regulated cell death, is primarily mediated by the activation of caspase proteases. Nevertheless, the resistance of many tumors to apoptotic pathways has shifted focus towards non-apoptotic forms of cell death, such as ferroptosis. Ferroptosis is an iron-dependent form of regulated necrosis characterized by extensive membrane damage resulting from lipid peroxidation. Numerous preclinical studies have demonstrated that inducing ferroptosis can significantly reduce tumor growth across a variety of cancer types. For instance, in a study involving breast cancer models, the use of ferroptosis inducers such as erastin and RSL3 led to a marked decrease in tumor volume and weight. This review aims to explore the potential of ferroptosis as a novel therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Oluwafemi Adeleke Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria; Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Susan Grant
- Department of Biochemistry, Landmark University, Omu-Aran, Nigeria
| | - Pearl Ifunanya Nwafor-Ezeh
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | | | | | - Emmanuel Henry Ezenabor
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | - Adebola Busola Ojo
- Department of Environmental Management and Toxicology, University of Ilesa, Ilesa, Nigeria
| |
Collapse
|
109
|
Banu H, Al-Shammari E, Shahanawaz S, Azam F, Patel M, Alarifi NA, Ahmad MF, Adnan M, Ashraf SA. Insights into the Therapeutic Targets and Molecular Mechanisms of Eruca sativa Against Colorectal Cancer: An Integrated Approach Combining Network Pharmacology, Molecular Docking and Dynamics Simulation. Pharmaceuticals (Basel) 2025; 18:453. [PMID: 40283891 PMCID: PMC12030293 DOI: 10.3390/ph18040453] [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: 02/03/2025] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: This study presents a novel and comprehensive investigation into the anti-colorectal cancer (CRC) effects and underlying mechanisms of Eruca sativa (E. sativa) using an integrated approach combining network pharmacology, molecular docking and molecular dynamics simulation. Methods: Using an integrated approach, six bioactive compounds and 40 potential targets were identified. A compound-target network was constructed, and enrichment analysis was performed to explore the key pathways influenced by E. sativa. Molecular docking analysis was used to evaluate the binding interactions between the identified compounds and key CRC-related targets (AKT1, PGR, MMP9, and PTGS2). Furthermore, molecular dynamics simulation was utilized to confirm the stability and reliability of these interactions. Results: The study found that E. sativa exhibits strong anticancer potential, particularly through major compounds such as β-ionone, 1-octanol, isorhamnetin, 2-hexenal, propionic acid, and quercetin. Molecular docking revealed favorable binding interactions between these compounds and key CRC targets, with quercetin and isorhamnetin showing the highest binding affinities. Additionally, molecular dynamics simulations validated the stability of these interactions, reinforcing their therapeutic relevance. Conclusions: This study provides valuable insights into the pharmacological mechanisms of E. sativa against CRC, highlighting its potential as a natural anticancer agent. These findings pave the way for future clinical studies to validate the efficacy and safety of E. sativa and its bioactive compounds, potentially contributing to the development of novel, plant-based therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Humera Banu
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| | - Eyad Al-Shammari
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| | - Syed Shahanawaz
- Department of Physiotherapy, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Faizul Azam
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Mitesh Patel
- Research and Development Cell, Parul University, Vadodara 391760, Gujarat, India;
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India
| | | | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia;
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| |
Collapse
|
110
|
Knight Z, Ruiz A, Elies J. Piezoelectric Nanomaterials for Cancer Therapy: Current Research and Future Perspectives on Glioblastoma. J Funct Biomater 2025; 16:114. [PMID: 40278222 PMCID: PMC12027790 DOI: 10.3390/jfb16040114] [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: 12/11/2024] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Cancer significantly impacts human quality of life and life expectancy, with an estimated 20 million new cases and 10 million cancer-related deaths worldwide every year. Standard treatments including chemotherapy, radiotherapy, and surgical removal, for aggressive cancers, such as glioblastoma, are often ineffective in late stages. Glioblastoma, for example, is known for its poor prognosis post-diagnosis, with a median survival time of approximately 15 months. Novel therapies using local electric fields have shown anti-tumour effects in glioblastoma by disrupting mitotic spindle assembly and inhibiting cell growth. However, constant application poses risks like patient burns. Wireless stimulation via piezoelectric nanomaterials offers a safer alternative, requiring ultrasound activation to induce therapeutic effects, such as altering voltage-gated ion channel conductance by depolarising membrane potentials. This review highlights the piezoelectric mechanism, drug delivery, ion channel activation, and current technologies in cancer therapy, emphasising the need for further research to address limitations like biocompatibility in whole systems. The goal is to underscore these areas to inspire new avenues of research and overcome barriers to developing piezoelectric nanoparticle-based cancer therapies.
Collapse
Affiliation(s)
- Zayne Knight
- Centre for Pharmaceutical Engineering Science, School of Pharmacy, University of Bradford, Bradford BD7 1DP, UK
| | - Amalia Ruiz
- Institute of Cancer Therapeutics (ICT), Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Jacobo Elies
- Institute of Cancer Therapeutics (ICT), Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
111
|
de Souza GA, Chaves LDS, Velez ASMM, Lacerda JLF, Pitasse-Santos P, dos Santos JCC, Chaves OA, Serpa C, Valente RDC, da Fonseca LM, da Costa Santos MAR, dos Reis JS, Santos CADN, Mendonça-Previato L, Previato JO, Freire-de-Lima CG, Decoté-Ricardo D, Freire-de-Lima L, de Lima MEF. Design and Synthesis of Bis-Chalcones as Curcumin Simplified Analogs and Assessment of Their Antiproliferative Activities Against Human Lung Cancer Cells and Trypanosoma cruzi Amastigotes. Pharmaceuticals (Basel) 2025; 18:456. [PMID: 40283893 PMCID: PMC12030033 DOI: 10.3390/ph18040456] [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: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Anticancer therapies represent the primary treatment option for a significant number of cancer patients globally; however, many of these treatments are associated with severe side effects as they target molecular structures present in both cancerous and healthy cells. In a similar context, the treatment of Chagas disease, a neglected tropical illness, is hindered by the high toxicity of the currently available drugs. Researchers are increasingly focusing on the development of safer and more selective alternatives, with natural compounds being studied as potential starting points for the creation of more effective drug candidates with a favorable therapeutic index. Objectives: The aim of this study was to design simplified curcumin-derived structures that preserved or enhanced their therapeutic activity against human lung cancer cell lines and T. cruzi, while also improving bioavailability and minimizing toxicity. Methods: In this study, curcumin and two natural curcuminoids inspired the synthesis of a chalcone and a set of bis-chalcones, compound classes known for their enhanced stability compared with their natural parent derivatives. The synthetic strategy used was the acid-catalyzed aldol condensation reaction. The stability profiles, IC50 values against A549 and H460 tumor cell lines, and trypanocidal activity against T. cruzi amastigotes of these derivatives were assessed. Results: The synthesized derivatives exhibited improved stability compared with the parent compounds, along with lower IC50 values in both A549 and H460 tumor cell lines. Additionally, one of the new analogs showed promising trypanocidal activity against T. cruzi amastigotes. Conclusions: This study provides a potential pathway toward the development of more effective and less toxic treatments for both cancer and Chagas disease. The simplified curcumin derivatives represent a promising foundation for designing new therapeutic agents with improved bioavailability and efficacy.
Collapse
Affiliation(s)
- Gabriela Alves de Souza
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Lorrane de Souza Chaves
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Afonso Santine M. M. Velez
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Jorge Lucas F. Lacerda
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Paulo Pitasse-Santos
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester LE1 7HB, UK;
- School of Chemistry, University of Leicester, Leicester LE1 7RH, UK
| | - Jayane Clys Conceição dos Santos
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Otávio Augusto Chaves
- Department of Chemistry, Coimbra Chemistry Centre-Institute of Molecular Science, University of Coimbra, 3004-535 Coimbra, Portugal; (O.A.C.); (C.S.)
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, RJ, Brazil
| | - Carlos Serpa
- Department of Chemistry, Coimbra Chemistry Centre-Institute of Molecular Science, University of Coimbra, 3004-535 Coimbra, Portugal; (O.A.C.); (C.S.)
| | - Raphael do Carmo Valente
- Campus Duque de Caxias Professor Geraldo Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25.240-005, RJ, Brazil;
| | - Leonardo Marques da Fonseca
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Curso de Medicina, Universidade Castelo Branco, Rio de Janeiro 21.710-255, RJ, Brazil
| | - Marcos André Rodrigues da Costa Santos
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Jhenifer Santos dos Reis
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Carlos Antônio do Nascimento Santos
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Lucia Mendonça-Previato
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Jose Osvaldo Previato
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Celio Geraldo Freire-de-Lima
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Debora Decoté-Ricardo
- Departamento de Microbiologia e Imunologia Veterinária, Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil;
| | - Leonardo Freire-de-Lima
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Programa de Pós-Graduação em Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Marco Edilson Freire de Lima
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| |
Collapse
|
112
|
Chen X. PDCA cycle and safety culture in nursing safety management of Day Ward chemotherapy. World J Surg Oncol 2025; 23:99. [PMID: 40121487 PMCID: PMC11929163 DOI: 10.1186/s12957-025-03738-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVE The PDCA cycle, also known as the PDCA (Plan-Do-Check-Act) cycle, is a well-established continuous quality improvement framework. This study aimed to evaluate the impact of implementing a nursing safety management strategy grounded in the PDCA cycle and safety culture principles in the context of Day Ward chemotherapy. METHODS This a prospective group comparison study (cohort comparison) based on principles of randomization. A total of 120 patients receiving intravenous chemotherapy at the Day Ward of Nantong First People's Hospital from January 2023 to December 2023 were recruited as research participants. They were randomly assigned to either a control group or a study group, which were managed using the conventional nursing quality management approach and the PDCA cycle-based safety culture management method, respectively. The primary outcomes measured were nursing satisfaction, chemotherapy-related symptom burden, and the incidence of total implantable venous access port catheter (TIVAP)-related adverse events. RESULTS After three months, the study group showed significantly lower scores on all MSAS-SF subscales (GDI: 1.05 ± 0.33, PHYS: 0.69 ± 0.35, PSYCH: 1.15 ± 0.42, TMSAS: 2.62 ± 0.34) compared to the control group (GDI: 1.22 ± 0.47, PHYS: 0.85 ± 0.32, PSYCH: 1.43 ± 0.73, TMSAS: 2.81 ± 0.36) (all P < 0.05). Nursing satisfaction was higher in the study group (95.00%) than in the control group (78.33%) (P < 0.05). Quality of life scores improved more in the study group (74.9 ± 9.2) than in the control group (68.2 ± 10.5) (P < 0.01). The study group also had fewer TIVAP-related adverse events (6.67%) compared to the control group (24.67%) (P < 0.05). CONCLUSION The adoption of a nursing safety management model rooted in the PDCA cycle and safety culture principles can effectively improve nursing quality and satisfaction, alleviate patient symptoms and enhance quality of life in the context of Day Ward chemotherapy. These findings underscore the merit of further disseminating and studying this management approach in nursing practice.
Collapse
Affiliation(s)
- Xia Chen
- Day Ward, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University (Nantong Hospital of Renji Hospital Afliated to Shanghai Jiao Tong University School of Medicine), No. 666 Shengli Road, Nantong City, Jiangsu, 226000, China.
- Nantong Hospital of Renji Hospital Afliated to Shanghai Jiao Tong University School of Medicine, Nantong City, Jiangsu, 226000, China.
| |
Collapse
|
113
|
Yean AW, Leong E, King OS, Mohamad Z. Breast cancer treatment modalities, treatment delays, and survival in Brunei Darussalam. BMC Cancer 2025; 25:510. [PMID: 40114099 PMCID: PMC11924827 DOI: 10.1186/s12885-025-13861-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
INTRODUCTION Breast cancer remains a leading cause of cancer-related mortality globally. This study aims to examine the demographic variables and effects of different treatment modalities and treatment delays on overall and relative survival rates of breast cancer patients in Brunei Darussalam. METHODS This retrospective study analysed data from the Brunei Darussalam Cancer Registry on breast cancer cases diagnosed and treated between 2013 and 2022. Statistical analyses included descriptive statistics to characterise the study population, Kaplan-Meier estimates to compare survival curves of different groups, Log rank tests to determine significant differences in survival rates among groups, and Cox Proportional Hazard (PH) models to estimate hazard ratios (HRs) and identify predictors of survival outcomes. Overall survival (OS) and relative survival (RS) rates were calculated. RESULTS Out of the 431 women treated for breast cancer, the majority were diagnosed at the regional stage (45.7%), with 39.0% at the localised stage. Over half (55.4%) of the diagnoses occurred in women aged 40 to 59, while about a quarter (25.5%) were in the 60-69 age group. Surgery was the most common first-line treatment modality (55.9%), with a median time to treatment of 37 days, followed by chemotherapy (30.6%). More than half of the patients (62.9%) were treated within 60 days of diagnosis. Treatment varied by age and cancer stage, with younger patients more likely to undergo surgery and older patients more likely to receive chemotherapy or hormonal therapy. Survival rates were high for patients receiving only surgery (5-year RS: 98.7%, OS: 92.3%), and significant survival differences were found for cancer stage and treatment delay, with a HR of 2.5 for delays over 60 days. Multivariate analysis showed that patients with distant stage cancer had a significantly higher risk of death (HR = 15.3) compared to localised stage. CONCLUSION This study highlights the impact of treatment modalities and delays on breast cancer survival in Brunei Darussalam, emphasising the need for timely treatment to improve survival rates. Our findings suggest that ensuring breast cancer treatment initiation within two months post-diagnosis may enhance patient outcomes, supporting potential policy targets for timely access to care.
Collapse
Affiliation(s)
- Ang Woan Yean
- Faculty of Science, Universiti Brunei Darussalam, Jln Tungku Link, Bandar Seri Begawan, Brunei Darussalam
| | - Elvynna Leong
- Faculty of Science, Universiti Brunei Darussalam, Jln Tungku Link, Bandar Seri Begawan, Brunei Darussalam.
| | - Ong Sok King
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jln Tungku Link, Bandar Seri Begawan, Brunei Darussalam
- Department of Policy and Planning, Ministry of Health, Bandar Seri Begawan, Brunei Darussalam
| | - Zulkhairi Mohamad
- The Brunei Cancer Centre, Jerudong Park Medical Centre, Jerudong, Bandar Seri Begawan, Brunei Darussalam
| |
Collapse
|
114
|
Lee CE, Noh KM, Kim S, Hong J, Kim K. Recent Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Engineering Strategies for Precise Strike Therapy against Tumor. Biomater Res 2025; 29:0170. [PMID: 40110051 PMCID: PMC11922527 DOI: 10.34133/bmr.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025] Open
Abstract
Effective drug delivery relies on the selection of suitable carriers, which is crucial for protein-based therapeutics such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). One of the key advantages of TRAIL is its ability to selectively induce apoptosis in cancer cells excluding healthy tissues by binding to death receptors DR4 and DR5, which are highly expressed in various cancer cells. Despite this promise, the clinical application of TRAIL has been limited by its short half-life, limited stability, and inefficient delivery to tumor sites. To overcome currently available clinical and engineering approaches, a series of sophisticated strategies is required: (a) the design of biomaterial-mediated carriers for enhanced targeting efficacy, particularly via optimizing selected materials, composition, formulation, and surface modulation. Moreover, (b) development of genetically modified cellular products for augmented TRAIL secretion toward tumor microenvironments and (c) cell surface engineering techniques for TRAIL immobilization onto infusible cell populations are also discussed in the present review. Among these approaches, living cell-based carriers offer the distinct advantage of systemically administered TRAIL-functionalized cells capturing circulating tumor cells in the bloodstream, thereby preventing secondary tumor formation. This review provides insight into the development of novel TRAIL delivery platforms, discusses considerations for clinical translation, and suggests future directions and complementary strategies to advance the field of TRAIL-based cancer therapeutics.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyung Mu Noh
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Jiyeon Hong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
115
|
Mukherjee A, Sarkar R. Unlocking the microbial treasure trove: advances in Streptomyces derived secondary metabolites in the battle against cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04001-5. [PMID: 40100372 DOI: 10.1007/s00210-025-04001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
Streptomyces is widely recognized as the "biological factory" of specialized metabolites comprising a huge variety of bioactive molecules with diverse chemical properties. The potential of this Gram-positive soil bacteria to produce such diversified secondary metabolites with significant biological properties positions them as an ideal candidate for anticancer drug discovery. Some of the Streptomyces-derived secondary metabolites include siderophores (enterobactin, desferrioxamine), antibiotics (xiakemycin, dinactin) pigments (prodigiosin, melanin), and enzymes (L-methioninase, L-asperginase, cholesterol oxidase) which exhibit a pronounced anticancer effect on both in vitro and in vivo system. These secondary metabolites are endowed with antiproliferative, pro-apoptotic, antimetastatic, and antiangiogenic properties, presenting several promising characteristics that make them suitable candidates in the battle against this deadly disease. In this comprehensive review, we have dived deep and explored their history of discovery, their role as anticancer agents, underlying mechanisms, the approaches for the discovery of anticancer molecules from the secondary metabolites of Streptomyces (isolation of Streptomyces, characterization of bacterial strain, screening for anticancer activity and determination of in vitro and in vivo toxicity, structure-activity relationship studies, clinical translation, and drug development studies). The hurdles and challenges associated with this process and their future prospect were also illustrated. This review highlights the efficacy of Streptomyces as a "microbial treasure island" for novel anticancer agents, which warrants sustained research and exploration in this field to disclose more molecules from Streptomyces that are unidentified and to translate the clinical application of these secondary metabolites for cancer patients.
Collapse
Affiliation(s)
- Adrija Mukherjee
- B.D. Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India
| | - Ruma Sarkar
- B.D. Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India.
| |
Collapse
|
116
|
Vu GTQ, Nguyen LM, Nguyen Do KN, Tran DL, Vo TV, Nguyen DH, Vong LB. Preparation of Metal-Polyphenol Modified Zeolitic Imidazolate Framework-8 Nanoparticles for Cancer Drug Delivery. ACS APPLIED BIO MATERIALS 2025; 8:2052-2064. [PMID: 39950754 DOI: 10.1021/acsabm.4c01608] [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: 02/18/2025]
Abstract
With the rising incidence of cancer, chemotherapy has become a widely used treatment approach. However, the use of anticancer drugs such as doxorubicin (DOX) poses significant long-term risks due to its nonspecific distribution and severe side effects. Therefore, developing a nanoparticle-based drug delivery system (DDS) that enhances the bioavailability of DOX specifically to cancer cells is crucial while minimizing its side effects on normal cells. This study employed zeolitic imidazolate framework-8 (ZIF-8) as a DDS to encapsulate DOX using a one-pot method. The surface of this system was subsequently modified with a copper-gallic acid (Cu-GA) complex to form the Cu-GA/DOX@ZIF-8 (CGDZ) system. The CGDZ system effectively encapsulates DOX and demonstrates pH-responsive drug release, facilitating controlled drug release in the acidic environment of cancer cells. Furthermore, the Cu-GA coating enhances the biocompatibility of the material, reduces drug toxicity in normal endothelial cells (BAECs) due to the antioxidant feature of modified GA, and maintains the efficacy and intracellular trafficking of DOX in colon cancer cells (C-26). Interestingly, CGDZ nanoparticles showed significantly higher toxicity against cancer cells as compared to unmodified systems and free DOX. Overall, CGDZ exhibited significant in vitro efficacy in targeting cancer cell lines while reducing the toxicity of DOX, offering a novel and effective nanoparticle system for targeted cancer treatment.
Collapse
Affiliation(s)
- Giao Thuy-Quynh Vu
- School of Biomedical Engineering, International University, Ho Chi Minh 700000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 700000, Vietnam
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, 1A TL29 Street, Thanh Loc Ward, District 12, Ho Chi Minh City 700000, Vietnam
| | - Luan Minh Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, 1A TL29 Street, Thanh Loc Ward, District 12, Ho Chi Minh City 700000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Kim Ngan Nguyen Do
- School of Biomedical Engineering, International University, Ho Chi Minh 700000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 700000, Vietnam
| | - Dieu Linh Tran
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, 1A TL29 Street, Thanh Loc Ward, District 12, Ho Chi Minh City 700000, Vietnam
| | - Toi Van Vo
- School of Biomedical Engineering, International University, Ho Chi Minh 700000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 700000, Vietnam
| | - Dai Hai Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology, 1A TL29 Street, Thanh Loc Ward, District 12, Ho Chi Minh City 700000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Street, Cau Giay District, Hanoi 100000, Vietnam
| | - Long Binh Vong
- School of Biomedical Engineering, International University, Ho Chi Minh 700000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 700000, Vietnam
| |
Collapse
|
117
|
Shaikh S, Chary PS, Mehra NK. Tyrosine Kinase Inhibitor Lenvatinib Based Nano Formulations and Cutting-Edge Scale-Up Technologies in revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1749-1784. [PMID: 40091597 DOI: 10.1021/acsabm.4c01527] [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: 03/19/2025]
Abstract
Lenvatinib (LEN), a tyrosine kinase inhibitor, has emerged as a promising therapeutic agent for various solid tumors. Nevertheless, a number of constraints, including diminished bioavailability, incapacity to elicit localized inflammation, and inability to selectively accumulate at the tumor site, may impede the comprehensive exploitation of its versatile tyrosine kinase inhibitory capabilities. In order to achieve targeted delivery of LEN while also reducing its high dose used in conventional therapeutics, nanoformulation approaches can be adopted. The integration of LEN into various nanoformulations, such as nanoparticles, nanocrystals, high density lipoproteins (HDLs), liposomes, and micelles, is discussed, highlighting the advantages of these innovative approaches in a comparative manner; however, given that the current methods of nanoformulation synthesis employ toxic organic solvents and chemicals, there is an imperative need for exploring alternative, environmentally friendly approaches. The multifaceted effects of nanocarriers have rendered them profoundly applicable within the biomedical domain, serving as instrumental entities in various capacities such as vehicles for drug delivery and genetic material, diagnostic agents, facilitators of photothermal therapy, and radiotherapy. However, the scalability of these nanotechnological methodologies must be rigorously investigated and addressed to refine drug delivery mechanisms. This endeavor offers promising prospects for revolutionizing strategies in cancer therapeutics, thereby laying the foundation for future research in scale-up techniques in the pursuit of more effective and less toxic therapies for cancer.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| |
Collapse
|
118
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
119
|
Gutiérrez Coronado O, Sandoval Salazar C, Muñoz Carrillo JL, Gutiérrez Villalobos OA, Miranda Beltrán MDLL, Soriano Hernández AD, Beltrán Campos V, Villalobos Gutiérrez PT. Functionalized Nanomaterials in Cancer Treatment: A Review. Int J Mol Sci 2025; 26:2633. [PMID: 40141274 PMCID: PMC11942109 DOI: 10.3390/ijms26062633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer is one of the main causes of death worldwide. Chemotherapy, radiotherapy and surgery are currently the treatments of choice for cancer. However, conventional therapies have their limitations, such as non-specificity, tumor recurrence and toxicity to the target cells. Recently, nanomaterials have been considered as therapeutic agents against cancer. This is mainly due to their unique optical properties, biocompatibility, large surface area and nanoscale size. These properties are crucial as they can affect biocompatibility and uptake by the cell, reducing efficacy. However, because nanoparticles can be functionalized with biomolecules, they become more biocompatible, which improves uptake, and they can be specifically targeted against cancer cells, which improves their anticancer activity. In this review, we summarize some of the recent studies in which nanomaterials have been functionalized with the aim of increasing therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Oscar Gutiérrez Coronado
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Mexico; (O.G.C.); (M.d.l.L.M.B.); (A.D.S.H.)
| | - Cuauhtémoc Sandoval Salazar
- División de Ciencias de la Salud e Ingenierías, Campus Celaya-Salvatierra, Universidad de Guanajuato, Celaya 38060, Mexico; (C.S.S.); (V.B.C.)
| | - José Luis Muñoz Carrillo
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Mexico; (O.G.C.); (M.d.l.L.M.B.); (A.D.S.H.)
| | | | - María de la Luz Miranda Beltrán
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Mexico; (O.G.C.); (M.d.l.L.M.B.); (A.D.S.H.)
| | | | - Vicente Beltrán Campos
- División de Ciencias de la Salud e Ingenierías, Campus Celaya-Salvatierra, Universidad de Guanajuato, Celaya 38060, Mexico; (C.S.S.); (V.B.C.)
| | | |
Collapse
|
120
|
Mohamed HRH, Elberry YA, Magdy H, Ismail M, Michael M, Eltayeb N, Safwat G. Erbium oxide nanoparticles induce potent cell death, genomic instability and ROS-mitochondrial dysfunction-mediated apoptosis in U937 lymphoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03962-x. [PMID: 40072553 DOI: 10.1007/s00210-025-03962-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Erbium oxide nanoparticles (Er2O3-NPs) have attracted significant attention for their unique physicochemical properties, including high surface area, biocompatibility, and stability. However, the impact of Er2O3-NPs on lymphoma cells (LCs) has not been explored, making this an innovative avenue for exploration. Therefore, the current study aimed to explore the influence of Er2O3-NPs on cell viability, genomic and mitochondrial DNA integrity, reactive oxygen species (ROS) generation and apoptosis induction in human U937 LCs. Er2O3-NPs were characterized using X-ray diffraction (XRD) and transmission electron microscopy (TEM). The effect of Er2O3-NPs on cell viability and genomic DNA integrity was estimated after 48 h using the WST-1 cytotoxicity and alkaline Comet assays, respectively. The generation level of reactive oxygen species (ROS) and mitochondrial membrane potential were also analyzed. Flow Cytometry was used to assess apoptosis induction and quantitative RT-PCR was conducted to measure the apoptotic (p53), anti-apoptotic (Bcl2), and mitochondrial (ND3) gene expression. Our results demonstrated the purity and well distribution of Er2O3-NPs and revealed that Er2O3-NPs induce strong cytotoxicity on U937 cells, as evidenced by a concentration-dependent reduction in cell viability with an IC50 value of 3.20 µg/ml. Exposure of U937 LCs to the IC50 concentration (3.20 µg/ml) of Er2O3-NPs promoted excessive ROS generation, leading to dramatic damage to genomic DNA and mitochondrial membrane potential, as well as marked dysregulation of apoptotic (p53), anti-apoptotic (Bcl2) and mitochondrial ND3 gene expression. This cascade of events triggered both apoptosis and necrosis in Er2O3-NPs-treated U937 LCs. In conclusion, these findings highlight the strong in vitro cytotoxic potential of Er2O3-NPs against highly aggressive U937 LCs, mediated by excessive ROS production, which leads to severe genomic DNA and mitochondrial membrane damage, as well as profound alterations in apoptotic, anti-apoptotic and mitochondrial gene expression. Future research is needed to further investigate the potential use of Er2O3-NPs in treating lymphoma and to optimize their therapeutic efficacy.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Yusuf Ahmed Elberry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Hagar Magdy
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Maryam Ismail
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Maivel Michael
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Nourhan Eltayeb
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| |
Collapse
|
121
|
Alwaili MA, Abu-Almakarem AS, El-Said KS, Eid TM, Mobasher MA, Alsabban AH, Alburae NA, Banjabi AA, Soliman MM. Shikimic acid protects against doxorubicin-induced cardiotoxicity in rats. Sci Rep 2025; 15:8126. [PMID: 40057537 PMCID: PMC11890735 DOI: 10.1038/s41598-025-90549-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
Doxorubicin (DOX) is used to treat a variety of malignancies; however, its cardiotoxicity limits its effectiveness. Shikimic acid (SA) showed several promising biomedical applications. This study investigated the protective effect of SA on DOX-induced cardiotoxicity in male rats. The ADMETlab 2.0 web server was used to predict the pharmacokinetic properties of SA. Molecular docking studies were conducted using AutoDock Vina. Fifty male rats were divided into 4 groups (n = 10); G1 was a negative control; G2 was injected with 4 mg/kg of DOX intraperitoneally (i.p.) once a week for a month; G3 was gavaged by 1/10 of SA LD50 (280 mg/kg) daily for a month, and G4 was injected with DOX as in G2 and with SA as in G3. After a month, hematological, biochemical, molecular, and histopathological investigations were assessed. The results showed that SA treatment led to significant amelioration of the DOX-induced cardiotoxicity in rats by restoring hematological, biochemical, inflammatory biomarkers, antioxidant gene expression, and cardiac histopathological alterations. Importantly, the impact of SA treatment against DOX-promoted cardiac deterioration is by targeting the Nrf-2/Keap-1/HO-1/NQO-1 signaling pathway, which in turn induces the antioxidant agents. These findings suggest that SA treatment could potentially mitigate cardiac toxicity during DOX-based chemotherapy.
Collapse
Affiliation(s)
- Maha Abdullah Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 11671, Riyadh, Saudi Arabia
| | - Amal S Abu-Almakarem
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
- Biochemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Maysa A Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, 72388, Sakaka, Saudi Arabia
| | - Ashwaq Hassan Alsabban
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Unit of Neurological Disorders, Department of Genetic Medicine, Faculty of Medicine, Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER.HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Najla Ali Alburae
- Department of Biological Science, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abeer A Banjabi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Mohamed Mostafa Soliman
- Department of Biology, College of Science, Jazan University, Kingdom of Saudi Arabia, P.O. Box. 114, 45142, Jazan, Saudi Arabia
- Department of Zoology, Faculty of Science, Ain Shams University, PO Box 11566, Cairo, Egypt
| |
Collapse
|
122
|
Yanova M, Stepanova E, Maltseva D, Tonevitsky A. CD44 variant exons induce chemoresistance by modulating cell death pathways. Front Cell Dev Biol 2025; 13:1508577. [PMID: 40114966 PMCID: PMC11924683 DOI: 10.3389/fcell.2025.1508577] [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: 10/10/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer chemoresistance presents a challenge in oncology, often leading to treatment failure and disease progression. CD44, a multifunctional cell surface glycoprotein, has garnered attention for its involvement in various aspects of cancer biology. Through alternative splicing, CD44 can form isoforms with the inclusion of only standard exons, typical for normal tissue, or with the addition of variant exons, frequently expressed in cancer tissue and associated with chemoresistance. The functions of CD44 involved in regulation of cancer signaling pathways are being actively studied, and the significance of specific variant exons in modulating cell death pathways, central to the response of cancer cells to chemotherapy, begins to become apparent. This review provides a comprehensive analysis of the association of CD44 variant exons/total CD44 with clinical outcomes of patients undergoing chemotherapy. The role of CD44 variant exons v6, v9 and others with a significant effect on patient chemotherapy outcomes by means of key cellular death pathways such as apoptosis, ferroptosis and autophagy modulation is further identified, and their impact on drug resistance is highlighted. An overview of clinical trials aimed at targeting variant exon-containing isoforms is provided, and possible directions for further development of CD44-targeted therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Maria Yanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Evgeniya Stepanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
123
|
Eiamart W, Wonganan P, Tadtong S, Samee W. Panduratin A from Boesenbergia rotunda Effectively Inhibits EGFR/STAT3/Akt Signaling Pathways, Inducing Apoptosis in NSCLC Cells with Wild-Type and T790M Mutations in EGFR. Int J Mol Sci 2025; 26:2350. [PMID: 40076971 PMCID: PMC11900324 DOI: 10.3390/ijms26052350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is a challenging disease, with the epidermal growth factor receptor (EGFR) being a key target for new, effective treatments crucial for the signaling pathways regulating cancer cell survival. Targeting EGFR-mediated signaling offers promising strategies to improve NSCLC therapies, particularly in overcoming resistance in EGFR-mutant lung cancer. In this study, we investigated the anticancer effects of panduratin A, a naturally occurring flavonoid from Boesenbergia rotunda, on human NSCLC cell lines expressing both wild-type EGFR (A549) and mutant EGFR (H1975) using in vitro experiments and molecular docking approaches. Cytotoxicity screening revealed that panduratin A exhibits potent effects on both A549 (IC50 of 6.03 ± 0.21 µg/mL) and H1975 (IC50 of 5.58 ± 0.15 µg/mL) cell lines while demonstrating low toxicity to normal MRC5 lung cells (12.96 ± 0.36 µg/mL). Furthermore, western blotting and flow cytometric analyses indicated that panduratin A induces apoptosis by inhibiting p-EGFR and its downstream effectors, p-STAT3 and p-Akt, in lung cancer cells. Additionally, the docking study showed lower binding energy between panduratin A and the target proteins, comparable to that of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs). The ADMET prediction also highlighted panduratin A's exceptional drug-like properties. This study concludes that panduratin A shows significant promise as an anti-lung cancer candidate for NSCLC, offering an economical and effective strategy.
Collapse
Affiliation(s)
- Wanna Eiamart
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Srinakharinwirot University, Nakhon Nayok 26120, Thailand;
- Chula Pharmacokinetic Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Piyanuch Wonganan
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Sarin Tadtong
- Department of Pharmacognosy, Faculty of Pharmacy, Srinakharinwirot University, Nakhon Nayok 26120, Thailand;
| | - Weerasak Samee
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Srinakharinwirot University, Nakhon Nayok 26120, Thailand;
| |
Collapse
|
124
|
Rizwan A, Sridharan B, Park JH, Kim D, Vial JC, Kyhm K, Lim HG. Nanophotonic-enhanced photoacoustic imaging for brain tumor detection. J Nanobiotechnology 2025; 23:170. [PMID: 40045308 PMCID: PMC11881315 DOI: 10.1186/s12951-025-03204-5] [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: 10/16/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Photoacoustic brain imaging (PABI) has emerged as a promising biomedical imaging modality, combining high contrast of optical imaging with deep tissue penetration of ultrasound imaging. This review explores the application of photoacoustic imaging in brain tumor imaging, highlighting the synergy between nanomaterials and state of the art optical techniques to achieve high-resolution imaging of deeper brain tissues. PABI leverages the photoacoustic effect, where absorbed light energy causes thermoelastic expansion, generating ultrasound waves that are detected and converted into images. This technique enables precise diagnosis, therapy monitoring, and enhanced clinical screening, specifically in the management of complex diseases such as breast cancer, lymphatic disorder, and neurological conditions. Despite integration of photoacoustic agents and ultrasound radiation, providing a comprehensive overview of current methodologies, major obstacles in brain tumor treatment, and future directions for improving diagnostic and therapeutic outcomes. The review underscores the significance of PABI as a robust research tool and medical method, with the potential to revolutionize brain disease diagnosis and treatment.
Collapse
Affiliation(s)
- Ali Rizwan
- Smart Gym-Based Translational Research Center for Active Senior'S Healthcare, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Badrinathan Sridharan
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Jin Hyeong Park
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Daehun Kim
- Indusrty 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Jean-Claude Vial
- Université Grenoble Alpes, CNRS, LIPhy, 38000, Grenoble, France
- Department of Optics & Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Kwangseuk Kyhm
- Department of Optics & Cogno-Mechatronics Engineering, Pusan National University, Busan, 46241, Republic of Korea
| | - Hae Gyun Lim
- Smart Gym-Based Translational Research Center for Active Senior'S Healthcare, Pukyong National University, Busan, 48513, Republic of Korea.
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
- Indusrty 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
125
|
Kenneth MJ, Wu CC, Fang CY, Hsu TK, Lin IC, Huang SW, Chiu YC, Hsu BM. Exploring the Impact of Chemotherapy on the Emergence of Antibiotic Resistance in the Gut Microbiota of Colorectal Cancer Patients. Antibiotics (Basel) 2025; 14:264. [PMID: 40149075 PMCID: PMC11939702 DOI: 10.3390/antibiotics14030264] [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: 01/13/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
With nearly half of colorectal cancer (CRC) patients diagnosed at advanced stages where surgery alone is insufficient, chemotherapy remains a cornerstone for this cancer treatment. To prevent infections and improve outcomes, antibiotics are often co-administered. However, chemotherapeutic interactions with the gut microbiota cause significant non-selective toxicity, affecting not only tumor and normal epithelial cells but also the gut microbiota. This toxicity triggers the bacterial SOS response and loss of microbial diversity, leading to bacterial mutations and dysbiosis. Consequently, pathogenic overgrowth and systemic infections increase, necessitating broad-spectrum antibiotics intervention. This review underscores how prolonged antibiotic use during chemotherapy, combined with chemotherapy-induced bacterial mutations, creates selective pressures that drive de novo antimicrobial resistance (AMR), allowing resistant bacteria to dominate the gut. This compromises the treatment efficacy and elevates the mortality risk. Restoring gut microbial diversity may mitigate chemotherapy-induced toxicity and improve therapeutic outcomes, and emerging strategies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, show considerable promise. Given the global threat posed by antibiotic resistance to cancer treatment, prioritizing antimicrobial stewardship is essential for optimizing antibiotic use and preventing resistance in CRC patients undergoing chemotherapy. Future research should aim to minimize chemotherapy's impact on the gut microbiota and develop targeted interventions to restore microbial diversity affected during chemotherapy.
Collapse
Affiliation(s)
- Mutebi John Kenneth
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Doctoral Program in Science, Technology, Environment and Mathematics, National Chung Cheng University, Chiayi 621, Taiwan
| | - Chin-Chia Wu
- Division of Colorectal Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
- College of Medicine, Tzu Chi University, Hualien 970, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chuan-Yin Fang
- Division of Colon and Rectal Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Tsui-Kang Hsu
- Department of Ophthalmology, Cheng Hsin General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - I-Ching Lin
- Department of Family Medicine, Asia University Hospital, Taichung 413, Taiwan
- Department of Kinesiology, Health and Leisure, Chienkuo Technology University, Changhua 500, Taiwan
| | - Shih-Wei Huang
- Center for Environmental Toxin and Emerging Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Yi-Chou Chiu
- General Surgery, Surgical Department, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Bing-Mu Hsu
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| |
Collapse
|
126
|
Zhong P, Li X, Li J. Mechanisms, assessment, and exercise interventions for skeletal muscle dysfunction post-chemotherapy in breast cancer: from inflammation factors to clinical practice. Front Oncol 2025; 15:1551561. [PMID: 40104495 PMCID: PMC11913840 DOI: 10.3389/fonc.2025.1551561] [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: 12/25/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Chemotherapy remains a central component of breast cancer treatment, significantly improving patient survival rates. However, its toxic side effects, along with cancer-related paraneoplastic syndromes, can lead to the loss of skeletal muscle mass and function, impairing physical abilities and increasing the risk of complications during treatment. Chemotherapeutic agents directly impact skeletal muscle cells by promoting protein degradation, inhibiting protein synthesis, and triggering systemic inflammation, all of which contribute to muscle atrophy. Additionally, these drugs can interfere with the proliferation and differentiation of stem cells, such as satellite cells, disrupting muscle regeneration and repair while inducing abnormal differentiation of intermuscular tissue, thereby worsening muscle wasting. These effects not only reduce the effectiveness of chemotherapy but also negatively affect patients' quality of life and disease prognosis. Recent studies have emphasized the role of exercise as an effective non-pharmacological strategy for preventing muscle loss and preserving muscle mass in cancer patients. This review examines the clinical manifestations of muscle dysfunction following breast cancer chemotherapy, the potential mechanisms underlying these changes, and the evidence supporting exercise as a therapeutic approach for improving muscle function.
Collapse
Affiliation(s)
- Pei Zhong
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xizhuang Li
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiehua Li
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
127
|
Liu H, Wang H, Lin X, Xu M, Lan W, Wang J. Harnessing natural saponins: Advancements in mitochondrial dysfunction and therapeutic applications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156383. [PMID: 39848019 DOI: 10.1016/j.phymed.2025.156383] [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: 11/06/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Mitochondrial dysfunction plays a crucial role in the development of a variety of diseases, notably neurodegenerative disorders, cardiovascular diseases, metabolic syndrome, and cancer. Natural saponins, which are intricate glycosides characterized by steroidal or triterpenoid structures, have attracted interest due to their diverse pharmacological benefits, including anti-inflammatory, antiviral, and anti-aging effects. PURPOSE This review synthesizes recent advancements in understanding mitochondrial dysfunction and explores how saponins can modulate mitochondrial function. It focuses on their potential applications in neuroprotection, cardiovascular health, and oncology. STUDY DESIGN The review incorporates a comprehensive literature analysis, highlighting the interplay between saponins and mitochondrial signaling pathways. Specific attention is given to the effects of saponins like ginsenoside Rg2 and 20(S)-protopanaxatriol on mitophagy and their neuroprotective, anti-aging, and synergistic therapeutic effects when combined. METHODS We conducted a comprehensive review of current research and clinical trials using PubMed, Google Scholar, and SciFinder databases. The search focused on saponins' role in mitochondrial function and their therapeutic effects, including "saponins", "mitochondria" and "mitochondrial function". The analysis primarily focused on articles published between 2011 and 2024. RESULTS The findings indicate that certain saponins can enhance mitophagy and modulate mitochondrial signaling pathways, showing promise in neuroprotection and anti-aging. Additionally, combinations of saponins have demonstrated synergistic effects in myocardial protection and cancer therapy, potentially improving therapeutic outcomes. CONCLUSION Although saponins exhibit significant potential in modulating mitochondrial functions and developing innovative therapeutic strategies, their clinical applications are constrained by low bioavailability. Rigorous clinical trials are essential to translate these findings into effective clinical therapies, ultimately improving patient outcomes through a deeper understanding of saponins' impact on mitochondrial function.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyu Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Min Xu
- Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Department of pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610000, China
| | - Wenying Lan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinlian Wang
- Traditional Chinese Medicine Hospital of Meishan, Meishan 620010, China.
| |
Collapse
|
128
|
Rijia A, Krishnamoorthi R, Mahalingam PU, Kaviyadharshini M, Rajeswari M, Kumar KKS, Rasmi M, Chung YK, Fang JY. Unveiling the anticancer potential and toxicity of Ganoderma applanatum wild mushroom derived bioactive compounds: An in vitro, in vivo and in silico evaluation. Bioorg Chem 2025; 156:108233. [PMID: 39908734 DOI: 10.1016/j.bioorg.2025.108233] [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: 01/06/2025] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
This study explores the anticancer potential of methanolic extract from Ganoderma applanatum, focusing on its cytotoxicity across various cancer cell lines and its safety and efficacy in an in vivo hepatocellular carcinoma (HCC) model, along with molecular docking analysis of its bioactive compounds targeting B-cell lymphoma 2 (Bcl-2) protein. The MTT assay revealed significant cytotoxicity of the extract against epidermoid carcinoma (A431), human alveolar carcinoma (A549), and hepatocellular carcinoma (HepG2) cell lines, with the extract exhibiting the highest potency (IC50 of 95.65 µg/ml) against HepG2 cells. Apoptosis induction and DNA degradation in HepG2 cells were confirmed through mitochondrial membrane potential analysis, ethidium bromide/acridine orange staining, and DNA fragmentation assays. In vivo studies on Wistar albino rats showed that administration of the extract up to 1000 mg/ml did not significantly affect body weight or hematological parameters, suggesting a favorable safety profile. Histopathological examination revealed normal liver architecture at most doses, with mild inflammation observed at the highest dose (1000 mg/ml). The G. applanatum extract were showed reducing liver weight and improving body weight in a Diethylnitrosamine (DEN)-induced HCC model was comparable to cyclophosphamide, indicating its potential as a less toxic alternative or adjunct to conventional chemotherapy. Additionally, the extract reduced elevated serum liver enzymes, demonstrating hepatoprotective effects. Molecular docking of nine bioactive compounds from G. applanatum identified 2h-3,11c-(epoxymethano)phenanthro[10,1-bc]pyran as a promising candidate for further investigation. These findings suggest G. applanatum as a novel anticancer agent with the potential for natural, effective cancer therapy.
Collapse
Affiliation(s)
- Akbar Rijia
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Raman Krishnamoorthi
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.
| | - Pambayan Ulagan Mahalingam
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India.
| | | | - Murugan Rajeswari
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Konda Kannan Satheesh Kumar
- Department of Chemistry, The Gandhigram Rural Institute (Deemed to be University), Gandhigram-624302, Dindigul, Tamil Nadu, India
| | - Madhusoodhanan Rasmi
- Department of Microbiology, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Yu-Kuo Chung
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| |
Collapse
|
129
|
Bahmanpour S, Ameri N, Zareifard N, Karimi F. The Protective Effect of GnRH Agonist Triptorelin on the Histomorphometric Parameters of the Utero-ovarian Tissue in the Doxorubicin- and Cyclophosphamide-treated Mice. Cell Biochem Biophys 2025; 83:573-586. [PMID: 39244688 DOI: 10.1007/s12013-024-01487-3] [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] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
One of the common side effects of chemotherapy drugs is ovarian failure and uterine dysfunction, which can occur after the administration of doxorubicin and/or cyclophosphamide. In clinics, gonadotropin-releasing hormone agonists (GnRHa) are used to modulate the toxic effect of chemotherapy and intercept infertility with some controversy and limited histological knowledge. This study aimed to evaluate the serological and histological features of protective effects of triptorelin, (GnRHa), on utero-ovarian tissue in the mice treated with cyclophosphamide and/or doxorubicin. Forty-eight female BALB/c mice were randomly divided into 8 groups as follows: Group I: normal saline; Group II: triptorelin; Group III: cyclophosphamide; Group IV: doxorubicin; Group V: cyclophosphamide + doxorubicin; and Groups VI, VII, and VIII: after injection of cyclophosphamide, doxorubicin, or cyclophosphamide + doxorubicin, administration of triptorelin (1 mg/kg; intraperitoneally) for 15 consecutive days, respectively. On the 21st day, the ovaries and uterine horns were dissected and weighed. Then, tissue processing and staining were performed for further histological and stereological studies. Triptorelin treatment in the damaged groups significantly increased the number of primordial and pre-antral follicles and granulosa cells. It decreased the number of atretic follicles compared to cyclophosphamide and/or doxorubicin-treated groups (P < 0.05). Triptorelin also significantly improved the volume of the ovary, cortex, medulla, oocytes in the primordial and antral follicles, uterus, endometrium, myometrium, uterine glands, and endometrial blood vessels in the damaged groups (P < 0.05). Triptorelin treatment prevents the destructive effects of cyclophosphamide and/or doxorubicin on utero-ovarian tissue.
Collapse
Affiliation(s)
- Soghra Bahmanpour
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Ameri
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nehleh Zareifard
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Karimi
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
130
|
Erboz A, Kesekler E, Gentili PL, Uversky VN, Coskuner-Weber O. Electromagnetic radiation and biophoton emission in neuronal communication and neurodegenerative diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:87-99. [PMID: 39732343 DOI: 10.1016/j.pbiomolbio.2024.12.004] [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: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
The intersection of electromagnetic radiation and neuronal communication, focusing on the potential role of biophoton emission in brain function and neurodegenerative diseases is an emerging research area. Traditionally, it is believed that neurons encode and communicate information via electrochemical impulses, generating electromagnetic fields detectable by EEG and MEG. Recent discoveries indicate that neurons may also emit biophotons, suggesting an additional communication channel alongside the regular synaptic interactions. This dual signaling system is analyzed for its potential in synchronizing neuronal activity and improving information transfer, with implications for brain-like computing systems. The clinical relevance is explored through the lens of neurodegenerative diseases and intrinsically disordered proteins, where oxidative stress may alter biophoton emission, offering clues for pathological conditions, such as Alzheimer's and Parkinson's diseases. The potential therapeutic use of Low-Level Laser Therapy (LLLT) is also examined for its ability to modulate biophoton activity and mitigate oxidative stress, presenting new opportunities for treatment. Here, we invite further exploration into the intricate roles the electromagnetic phenomena play in brain function, potentially leading to breakthroughs in computational neuroscience and medical therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Aysin Erboz
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Elif Kesekler
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Pier Luigi Gentili
- Department of Chemistry, Biology, and Biotechnology, Università degli Studi di Perugia, 06123, Perugia, Italy.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL 33612, USA.
| | - Orkid Coskuner-Weber
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey.
| |
Collapse
|
131
|
Mohiuddin AKM, Ferdous N, Reza MN, Al Amin M, Khanam R, Hossain MU, Ahammad I, Mahmud S. Designing siRNA for silencing the human ERBB2 gene in cancer treatment: Evaluating intracellular delivery strategies. Comput Biol Med 2025; 186:109663. [PMID: 39809083 DOI: 10.1016/j.compbiomed.2025.109663] [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: 06/24/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
The ERBB2 is one of the most studied genes in oncology for its significant role in human malignancies. The metastasis-associated properties that facilitate cancer metastasis can be enhanced by activating the ERBB2 receptor signaling pathways. Additionally, therapeutic resistance is conferred by ERBB2 overexpression via receptor-mediated antiapoptotic signals. Several ERBB2-blocking techniques have the effect of overexpressed ERBB2, and several of them have passed clinical trials for use as therapies. Small interfering RNAs (siRNA), which have the potential to silence genes, are attractive for treating such fatal malignancies. In this study, we rationally designed a siRNA molecule targeting the human ERBB2 gene. The selection process involved identifying a shared region among all ERBB2 transcripts for siRNA design. The ultimate siRNA candidate was chosen through rigorous evaluation using contemporary algorithms, considering off-target similarities, examination of thermodynamic properties, and analysis using molecular dynamics (MD) simulations. Further, we opted for cell-penetrating peptides (CPP) and RNA aptamer as carriers for the siRNA. Employing both steered MD simulations and traditional MD simulations, we investigated how these carriers facilitate siRNA delivery. Experimental confirmation revealed the stability of the selected carriers and siRNA on the lipid bilayer. The designed siRNA molecule and the simulations present a potential alternative therapeutic strategy against human ERBB2. This contributes to advances in developing and utilizing innovative carriers for the delivery of siRNA, enhancing the potential for therapeutic applications.
Collapse
Affiliation(s)
- A K M Mohiuddin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh.
| | - Nadim Ferdous
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Mahjerin Nasrin Reza
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Md Al Amin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Roksana Khanam
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ashulia, Savar, Dhaka-1349, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| |
Collapse
|
132
|
Gupta U, Kavya MP, Jayalakshmi K, Sinha N. Unlocking the Chemical Diversity of Plant Catharanthus roseus: Nuclear Magnetic Resonance Spectroscopy Approach. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2025; 63:205-215. [PMID: 39702899 DOI: 10.1002/mrc.5504] [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: 09/10/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Catharanthus roseus, also known as Madagascar periwinkle, is a perennial plant renowned for its extensive pharmacological properties. It produces vital chemotherapeutic compounds, including vinblastine and vincristine, and exhibits anti-inflammatory, antidiabetic, and antioxidant activities. In this study, we utilized a range of two-dimensional (2D) nuclear magnetic resonance (NMR) techniques, such as 1H-1H correlation spectroscopy (COSY), 1H-1H J-resolved NMR, and 1H-13C heteronuclear single quantum coherence (HSQC) sensitivity-enhanced NMR spectroscopy, to identify key metabolites in C. roseus leaf extracts. Given the presence of numerous metabolites with closely spaced multiplet resonances, the 1H NMR spectra often exhibit significant signal overlap, making metabolite identification difficult or even impossible. However, the use of 2D NMR techniques effectively overcame this challenge, allowing for the precise identification of important alkaloids, such as vindoline, vinblastine, serpentine, and ajmalicine, along with essential metabolites like organic acids, amino acids, and carbohydrates. The extract contained a variety of bioactive compounds, including organic acids crucial for the tricarboxylic acid (TCA) cycle, branched-chain amino acids vital for metabolic functions, and alkaloids with substantial therapeutic potential. This comprehensive study underscores the continued significance of C. roseus in both traditional and modern medicine, emphasizing its intricate metabolic network and its potential in the development of novel therapeutics.
Collapse
Affiliation(s)
- Upasna Gupta
- Centre of Bio-Medical Research (CBMR), Sanjay Gandhi Post Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - M P Kavya
- Department of Chemistry, Karnatak University's Karnatak Science College, Dharwad, India
| | - K Jayalakshmi
- Department of Chemistry, Karnatak University's Karnatak Science College, Dharwad, India
| | - Neeraj Sinha
- Centre of Bio-Medical Research (CBMR), Sanjay Gandhi Post Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
133
|
Choi YH, Kim HY, Park JO, Choi E. Enhanced Anti-Tumor Effects of Natural Killer Cell-Derived Exosomes Through Doxorubicin Delivery to Hepatocellular Carcinoma Cells: Cytotoxicity and Apoptosis Study. Int J Mol Sci 2025; 26:2234. [PMID: 40076856 PMCID: PMC11900065 DOI: 10.3390/ijms26052234] [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: 01/17/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Exosomes are nanosized extracellular vesicles secreted by various cells, including natural killer (NK) cells, and are known for their low toxicity, high permeability, biocompatibility, and strong targeting ability. NK cell-derived exosomes (NK-exos) contain cytotoxic proteins that enhance tumor-targeting efficiency, making them suitable for treating solid tumors such as hepatocellular carcinoma (HCC). Despite their potential in drug delivery, the mechanisms of drug-loaded NK-exos, particularly those loaded with doxorubicin (NK-exos-Dox), remain unclear in HCC. This study explored the anti-tumor effects of NK-exos-Dox against Hep3B cells in vitro. NK-exos-Dox expressed exosome markers (CD9 and CD63) and cytotoxic proteins (granzyme B and perforin) and measured 170-220 nm in size. Compared to NK-exos, NK-exos-Dox enhanced cytotoxicity and apoptosis in Hep3B cells by upregulating pro-apoptotic proteins (Bax, cytochrome c, cleaved caspase 3, and cleaved PARP) and inhibiting the anti-apoptotic protein (Bcl-2). These findings suggest that NK-exos-Dox significantly boost anti-tumor effects by activating specific cytotoxic molecules, offering promising therapeutic opportunities for solid tumor treatment, including HCC.
Collapse
Affiliation(s)
- You Hee Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Eunpyo Choi
- Department of Mechanical Engineering, Sogang University, 35, Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
134
|
Nainggolan SI, Rajuddin R, Kamarlis RK, Hambal M, Frengki F. In silico study of the potential of curcumin and its derivatives for increasing wild-type p53 expression and improving the function of p53 mutant R273H. Vet World 2025; 18:715-730. [PMID: 40342745 PMCID: PMC12056914 DOI: 10.14202/vetworld.2025.715-730] [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: 10/20/2024] [Accepted: 02/25/2025] [Indexed: 05/11/2025] Open
Abstract
Background and Aim p53 is a critical tumor suppressor protein responsible for regulating the cell cycle and inducing apoptosis. Mutations in the p53 gene, particularly in the DNA-binding domain, are frequently associated with various cancers due to the loss of transcriptional activity. Curcumin and its derivatives have demonstrated potential as p53 enhancers and reactivators of mutant p53. This study employs in silico methods to evaluate the potential of curcumin derivatives to enhance wild-type p53 expression and restore the function of the p53 mutant R273H. Materials and Methods Curcumin and 20 derivatives were selected from PubChem for computational analysis. Their potential as p53 enhancers was assessed using Quantitative Structure-Activity Relationship (QSAR) analysis. Molecular docking was conducted to determine their binding affinities with wild-type and mutant p53 proteins, followed by molecular dynamics (MD) simulations to evaluate ligand-receptor stability. Pharmacokinetics and toxicity assessments were performed using predictive computational models to evaluate their drug-like properties. Results QSAR analysis identified hexahydrocurcumin (probable activity [Pa]: 0.837) and tetrahydrocurcumin (Pa: 0.752) as the most potent p53 enhancers. Molecular docking revealed strong binding affinities for curcumin derivatives at key p53 binding residues, particularly through hydrogen bonds with His 273 of the R273H mutant. MD simulations demonstrated that curcumin, bisdemethoxycurcumin, and monodemethylcurcumin stabilized p53 mutant R273H, closely mimicking the structural stability of wild-type p53. Pharmacokinetic analysis indicated favorable absorption, distribution, metabolism, and excretion profiles for most derivatives, with low toxicity predicted for the majority. Conclusion Curcumin and its derivatives exhibit dual functions as p53 enhancers and reactivators of the p53 mutant R273H. Hexahydrocurcumin and tetrahydrocurcumin emerged as promising compounds with strong bioactivity and favorable pharmacokinetic properties, suggesting their potential as anticancer agents. Further in vitro and in vivo studies are necessary to validate these findings and explore their therapeutic applications.
Collapse
Affiliation(s)
- Sarah Ika Nainggolan
- Graduate School of Mathematics and Applied Sciences, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
- Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology, Faculty of Medicine, Syiah Kuala University, Banda Aceh, Indonesia
| | - Rajuddin Rajuddin
- Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology, Faculty of Medicine, Syiah Kuala University, Banda Aceh, Indonesia
| | - Reno Keumalazia Kamarlis
- Department of Obstetrics and Gynecology, Division of Gynecological Endocrinology, Faculty of Medicine, Syiah Kuala University, Banda Aceh, Indonesia
| | - Muhammad Hambal
- Department of Parasitology, Faculty of Medicine Veterinary, Syiah Kuala University, Banda Aceh, 243111, Indonesia
| | - Frengki Frengki
- Department of Pharmacology, Faculty of Medicine Veterinary, Syiah Kuala University, Banda Aceh, Indonesia
| |
Collapse
|
135
|
Thakur GS, Gupta AK, Pal D, Vaishnav Y, Kumar N, Annadurai S, Jain SK. Designing novel cabozantinib analogues as p-glycoprotein inhibitors to target cancer cell resistance using molecular docking study, ADMET screening, bioisosteric approach, and molecular dynamics simulations. Front Chem 2025; 13:1543075. [PMID: 40084274 PMCID: PMC11903459 DOI: 10.3389/fchem.2025.1543075] [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: 12/10/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction One of the foremost contributors to mortality worldwide is cancer. Chemotherapy remains the principal strategy for cancer treatment. A significant factor leading to the failure of cancer chemotherapy is the phenomenon of multidrug resistance (MDR) in cancer cells. The primary instigator of MDR is the over expression of P-glycoprotein (P-gp), a protein that imparts resistance and facilitates the ATP-dependent efflux of various anticancer agents. Numerous efforts have been made to inhibit P-gp function with the aim of restoring the effectiveness of chemotherapy due to its broad specificity. The main objective has been to create compounds that either serve as direct P-gp inhibitors or interact with cancer therapies to modulate transport. Despite substantial in vitro achievements, there are currently no approved drugs available that can effectively "block" P-gp mediated resistance. Cabozantinib (CBZ), a multi-kinase inhibitor, is utilized in the treatment of various carcinomas. CBZ has been shown to inhibit P-gp efflux activity, thereby reversing P-gp mediated MDR. Consequently, P-gp has emerged as a critical target for research in anti-cancer therapies. Methods The purpose of this study was to computationally identify new andsafer analogues of CBZ using bioisosteric approach, focusing on improved pharmacokinetic properties andreduced toxicity. The physicochemical, medicinal, and ADMET profiles of generated analogues were computed using the ADMETLab 3.0 server. We also predicted the drug likeness (DL) and drug score (DS) of analogues. The molecular docking studies of screened analogues against the protein (PDB ID: 3G5U) were conducted using AutoDock Vina flowing by BIOVIA Discovery Studio for visualizing interactions.Molecular dynamics (MD) simulation of docked ligands was done using Schrödinger suite. Results and Discussion The docking scores for the ligands CBZ01, CBZ06, CBZ11, CBZ13, CBZ25, CBZ34, and CBZ38 ranged from -8.0 to -6.4 kcal/mol against the protein (PDB ID: 3G5U). A molecular dynamics (MD) simulation of CBZ01, CBZ13, and CBZ38 was conducted using the Schrödinger suite, revealing that these complexesmaintained stability throughout the 100 ns simulation. Conclusion An integrated computational approach combining bioisosteric approach, molecular docking, drug likeness calculations, and MD simulations highlights the promise of ligands CBZ01 and CBZ13 as candidates for the development of potential anticancer agents for the treatment of various cancers.
Collapse
Affiliation(s)
- Gajendra Singh Thakur
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ajay Kumar Gupta
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Dipti Pal
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Yogesh Vaishnav
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles’ College of Pharmacy, Udaipur, Rajasthan, India
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sanmati Kumar Jain
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
136
|
Rangel-López R, Franco-Molina MÁ, Rodríguez-Padilla C, Zárate-Triviño DG. Gold Nanoparticles Synthesized with Triple-Negative Breast Cancer Cell Lysate Enhance Antitumoral Immunity: A Novel Synthesis Method. Pharmaceuticals (Basel) 2025; 18:330. [PMID: 40143109 PMCID: PMC11945454 DOI: 10.3390/ph18030330] [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: 01/09/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Gold nanoparticles enhance immunity, promotes antigen uptake by antigen-presenting cells (APCs), and boost the response against tumor antigens; therefore, they are a promising delivery vehicle. Tumor lysates have shown favorable responses as inductors of anti-cancer immunity, but the effectiveness of these treatments could be improved. Hybrid nanosystems gold nanoparticles with biomolecules have been show promising alternative on uptake, activation and response on immune system. Objectives: This study's objective was to develop a method of synthesizing gold nanoparticles employing a triple-negative breast cancer (4T1) cell lysate (AuLtNps) as a reducing agent to increase immunogenicity against breast cancer cells. Methods: Nanoparticle formation, size, and ζ potential were confirmed by surface plasmon resonance, dynamic light scattering, and transmission electron microscopy. Protein concentration was quantified using a Pierce BCA assay. The cytotoxic effects of treatments on murine macrophages were assessed, along with nanoparticle and tumor lysate uptake via epifluorescence microscopy. Using a murine model, cytokine secretion profiles were determined, and the efficacy in inhibiting the implantation of a 4T1 model was evaluated. Results/Conclusions: AuLtNps exhibited higher protein content than tumor lysate alone, leading to increased uptake and phagocytosis in murine macrophages, as confirmed by epifluorescence microscopy. Cytokine secretion analysis showed a proinflammatory response, with increased CD8+ and CD22+ lymphocytes and upregulation of APC markers (CD14, CD80, CD86, and MHC II+). Splenocytes demonstrated specific lysis of up to 40% against 4T1 tumor cells. In a murine model, AuLtNPs effectively inhibited tumor implantation, achieving an improved 90-days survival rate, highlighting their potential as an immunotherapy for triple-negative breast cancer.
Collapse
|
137
|
Ottaviani A, Pietrafesa D, Soren BC, Dasari JB, Olsen SSH, Messina B, Demofonti F, Chicarella G, Agama K, Pommier Y, Morozzo della Rocca B, Iacovelli F, Romeo A, Falconi M, Baker BJ, Fiorani P. Unveiling the Mechanism of Action of Palmitic Acid, a Human Topoisomerase 1B Inhibitor from the Antarctic Sponge Artemisina plumosa. Int J Mol Sci 2025; 26:2018. [PMID: 40076642 PMCID: PMC11900379 DOI: 10.3390/ijms26052018] [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: 01/24/2025] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a leading cause of death worldwide, highlighting the urgent need for novel and more effective treatments. Natural products, with their structural diversity, represent a valuable source for the discovery of anticancer compounds. In this study, we screened 750 Antarctic extracts to identify potential inhibitors of human topoisomerase 1 (hTOP1), a key enzyme in DNA replication and repair, and a target of cancer therapies. Bioassay-guided fractionation led to the identification of palmitic acid (PA) as the active compound from the Antarctic sponge Artemisina plumosa, selectively inhibiting hTOP1. Our results demonstrate that PA irreversibly blocks hTOP1-mediated DNA relaxation and specifically inhibits the DNA religation step of the enzyme's catalytic cycle. Unlike other fatty acids, PA exhibited unique specificity, which we confirmed through comparisons with linoleic acid. Molecular dynamics simulations and binding assays further suggest that PA interacts with hTOP1-DNA complexes, enhancing the inhibitory effect in the presence of camptothecin (CPT). These findings identify PA as a hTOP1 inhibitor with potential therapeutic implications, offering a distinct mechanism of action that could complement existing cancer therapies.
Collapse
Affiliation(s)
- Alessio Ottaviani
- Department of Onco-Hematology, Gene and Cell Therapy, Bambino Gesù Children’s Hospital-IRCCS, Via Ferdinando Baldelli 38, 00146 Rome, Italy;
| | - Davide Pietrafesa
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Bini Chhetri Soren
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Jagadish Babu Dasari
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Stine S. H. Olsen
- Department of Chemistry, University of South Florida, USF Sweetgum Ln 12111, Tampa, FL 33620, USA; (S.S.H.O.)
| | - Beatrice Messina
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Francesco Demofonti
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Giulia Chicarella
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Keli Agama
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Convent Drive 37, Bethesda, MD 20892, USA; (K.A.); (Y.P.)
| | - Yves Pommier
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Convent Drive 37, Bethesda, MD 20892, USA; (K.A.); (Y.P.)
| | - Blasco Morozzo della Rocca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Alice Romeo
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
| | - Bill J. Baker
- Department of Chemistry, University of South Florida, USF Sweetgum Ln 12111, Tampa, FL 33620, USA; (S.S.H.O.)
| | - Paola Fiorani
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; (D.P.); (B.C.S.); (J.B.D.); (B.M.); (F.D.); (G.C.); (B.M.d.R.); (F.I.); (A.R.); (M.F.)
- Institute of Translational Pharmacology, National Research Council, CNR, Via del Fosso del Cavaliere 100, 00133 Rome, Italy
| |
Collapse
|
138
|
Gonçalves E, Fontes F, Rodrigues JR, Antunes L, Bento MJ, Lunet N, Morais S. Temporal trends in the incidence of second primary cancers in Northern Portugal: a population-based study. Eur J Cancer Prev 2025:00008469-990000000-00212. [PMID: 40019750 DOI: 10.1097/cej.0000000000000962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The growing number of cancer survivors has led to an increase in the frequency of multiple primary cancers. This study aimed to describe the temporal trends in the incidence rates and standardized incidence ratios (SIRs) of second primary cancers (SPCs) among patients diagnosed with a first primary cancer (FPC) in 2000-03, 2004-07 and 2008-11 in Northern Portugal. Population-based samples of patients diagnosed with an FPC (excluding skin non-melanoma) in 3 periods of 4 years, between 1 January 2000 and 31 December 2011, and registered in the Portuguese North Region Cancer Registry were followed for the diagnosis of an SPC or death until 31 December 2013. Incidence rates and SIRs were estimated for each period by sex, and considering the time between FPC and SPC diagnosis (synchronous and metachronous). During follow-up, 10 119 (7.4%) SPCs were identified among patients diagnosed with an FPC in 2000-11 (n = 136 382). The incidence rate of SPCs was over 10-fold higher in the first few months, remaining stable over the follow-up. Higher rates were observed in 2004-07 and 2008-11. Overall, a greater proportion of SPCs was diagnosed among males than females. The proportion of SPCs diagnosed increased with age. Increases in SIRs of SPCs were observed from 2000-03 to 2008-11, ranging from 1.16 to 1.77 and from 1.54 to 2.33, among males and females, respectively. This was particularly evident for FPCs and SPCs of the lip, oral cavity, pharynx, oesophagus and larynx among males, and colon and rectum, lung, ovary and cervix among females. Survivors of cancer in Northern Portugal had higher incidence rates of cancer than the general population, which have increased over time. Our findings highlight the need for enhanced surveillance and tailored strategies for survivors of cancer, emphasizing the challenges of their heightened cancer risk, patient expectations, and associated economic burden.
Collapse
Affiliation(s)
- Elisabete Gonçalves
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR)
| | - Filipa Fontes
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto
- Grupo de Abordagem de Lesões Pré-Cancerosas e Cancro Precoce
| | - Jéssica Rocha Rodrigues
- Grupo de Investigação em Epidemiologia, Resultados, Economia e Gestão em Oncologia - Centro de Investigação (CI-IPOP) & Porto Comprehensive Cancer Center (Porto.CCC) & RISE@CI-IPOP (Rede de Investigação em Saúde)
- Serviço de Epidemiologia, Instituto Português de Oncologia do Porto FG, EPE (IPO-Porto)
| | - Luis Antunes
- Grupo de Investigação em Epidemiologia, Resultados, Economia e Gestão em Oncologia - Centro de Investigação (CI-IPOP) & Porto Comprehensive Cancer Center (Porto.CCC) & RISE@CI-IPOP (Rede de Investigação em Saúde)
| | - Maria José Bento
- Grupo de Investigação em Epidemiologia, Resultados, Economia e Gestão em Oncologia - Centro de Investigação (CI-IPOP) & Porto Comprehensive Cancer Center (Porto.CCC) & RISE@CI-IPOP (Rede de Investigação em Saúde)
- Serviço de Epidemiologia, Instituto Português de Oncologia do Porto FG, EPE (IPO-Porto)
- Departamento de Estudos de Populações, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Nuno Lunet
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR)
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina da Universidade do Porto
| | - Samantha Morais
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR)
| |
Collapse
|
139
|
Katabalo DM, Abraham M, Kidenya BR, Liwa A, Schroeder K. Prescription patterns of supportive care medications among children receiving chemotherapy treatments at a major referral hospital in Tanzania: where are we in managing chemotherapy-induced toxicities? Front Oncol 2025; 15:1444565. [PMID: 40071090 PMCID: PMC11893393 DOI: 10.3389/fonc.2025.1444565] [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: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Background Cancer chemotherapy is a treatment that systematically kills cancer cells but causes expected side effects, known as chemotherapy-induced toxicities. These toxicities are managed with supportive care medications. This study aimed to determine the prescription patterns of supportive care medications in children receiving chemotherapy at a major referral hospital in Tanzania. Methodology A hospital-based descriptive cross-sectional study was conducted at Bugando Medical Centre (BMC). The study analyzed 104 prescription slips of pediatric cancer patients receiving chemotherapy and qualitatively assessed national guidelines and disease-specific protocols used in guiding treatment. Data were cleaned in Microsoft Excel, analyzed using STATA version 15, and presented as frequencies, percentages, and narrative summaries. Results Ondansetron (84.6%) and pre-hydration normal saline (20.2%) were the most prescribed pre-chemotherapy supportive care medications. Similarly, oral ondansetron (80.8%) and post-hydration normal saline (22.1%) were the most prescribed post-chemotherapy medications. Few prescriptions included a combination of antiemetics, fluids, and proton pump inhibitors for regimens with multiple chemotherapeutic agents. National cancer treatment guidelines lacked detailed sections on supportive care medications, leaving prescribing decisions to clinicians, while Burkitt's lymphoma and nephroblastoma protocols offered more detailed guidance. Conclusion Antiemetics and hydration fluids dominated supportive care prescriptions. Significant gaps were identified in the inclusion of supportive care in national guidelines, with reliance on disease-specific protocols. These findings highlight the need for standardized, evidence-based supportive care guidelines tailored to resource-limited settings.
Collapse
Affiliation(s)
- Deogratias M. Katabalo
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
- Department of Oncology, Bugando Medical Centre, Mwanza, Tanzania
| | - Melina Abraham
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Benson R. Kidenya
- Department of Biochemistry, School of Medicine, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Antony Liwa
- Department of Pharmacology, School of Medicine, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Kristin Schroeder
- Department of Oncology, Bugando Medical Centre, Mwanza, Tanzania
- Duke Global Health Institute, Duke University, Durham, NC, United States
| |
Collapse
|
140
|
Choe S, Jeon M, Yoon H. Advanced Therapeutic Approaches for Metastatic Ovarian Cancer. Cancers (Basel) 2025; 17:788. [PMID: 40075635 PMCID: PMC11898553 DOI: 10.3390/cancers17050788] [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: 01/27/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death among women, which is one of the most common gynecological cancers worldwide. Although several cytoreductive surgeries and chemotherapies have been attempted to address ovarian cancer, the disease still shows poor prognosis and survival rates due to prevalent metastasis. Peritoneal metastasis is recognized as the primary route of metastatic progression in ovarian cancer. It causes severe symptoms in patients, but it is generally difficult to detect at an early stage. Current anti-cancer therapy is insufficient to completely treat metastatic ovarian cancer due to its high rates of recurrence and resistance. Therefore, developing strategies for treating metastatic ovarian cancer requires a deeper understanding of the tumor microenvironment (TME) and the identification of effective therapeutic targets through precision oncology. Given that various signaling pathways, including TGF-β, NF-κB, and PI3K/AKT/mTOR pathways, influence cancer progression, their activity and significance can vary depending on the cancer type. In ovarian cancer, these pathways are particularly important, as they not only drive tumor progression but also impact the TME, which contributes to the metastatic potential. The TME plays a critical role in driving metastatic features in ovarian cancer through altered immunologic interactions. Recent therapeutic advances have focused on targeting these distinct features to improve treatment outcomes. Deciphering the complex interaction between signaling pathways and immune populations contributing to metastatic ovarian cancer provides an opportunity to enhance anti-cancer efficacy. Hereby, this review highlights the mechanisms of signaling pathways in metastatic ovarian cancer and immunological interactions to understand improved immunotherapy against ovarian cancer.
Collapse
Affiliation(s)
- Soohyun Choe
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Minyeong Jeon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hyunho Yoon
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Republic of Korea; (S.C.); (M.J.)
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| |
Collapse
|
141
|
Rezaeianzadeh O, Asghari S, Tajbakhsh M, Khalilpour A, Shityakov S. Synthesis and application of diazenyl sulfonamide-based schiff bases as potential BRCA2 active inhibitors against MCF-7 breast cancer cell line. Sci Rep 2025; 15:6661. [PMID: 39994448 PMCID: PMC11850876 DOI: 10.1038/s41598-025-91113-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
In this study, a library of novel sulfonamide-based Schiff bases 3a-j was synthesized in high yield (75 to 89%). The FTIR, 1H NMR, and 13C NMR spectroscopic techniques and mass analysis were used to characterize the synthesized compounds. Their anticancer activity was assessed in vitro on the breast cancer (MCF-7) and healthy human breast epithelial (MCF-10 A) cell lines over 48 and 72 h using the MTT assay. Most of the synthesized compounds demonstrated promising activity, with compound 3i showing particularly high efficacy at 48 and 72 h (IC50 = 4.85 ± 0.006 and 4.25 ± 0.009 µM) against the MCF-7 breast cancer cell line. Furthermore, molecular docking studies were performed for compounds 3a-j with the PDB: (3UV7) protein of the breast cancer susceptibility gene 2 (BRCA2). The obtained results revealed that compound 3i has the strongest binding affinity energy (-7.99 kcal/mol), consistent with the obtained experimental data. Additionally, molecular dynamics (MD) simulation assays confirm the formation of a stable 3i-BRCA2 complex with strong binding affinity through the formation of hydrogen bonds. Antioxidant activities were determined by in vitro assay DPPH cation radical activity method. Interestingly, the compound 3j (IC50 = 12.36 ± 0.55 µM) had comparable activity with ascorbic acid (IC50 = 13.58 ± 0.38 µM) in the antioxidant assay. The results of this research could potentially contribute to the development of new therapeutic agents useful in fighting caused by breast cancer.
Collapse
Affiliation(s)
- Olia Rezaeianzadeh
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran
| | - Sakineh Asghari
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran.
| | - Mahmood Tajbakhsh
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran
| | - Asieh Khalilpour
- Department of Environmental Health Engineering, Babol University of Medicinal Sciences, Babol, Iran
| | - Sergey Shityakov
- Infochemistry Scientific Center, ITMO University, Lomonosova str. 9, 191002, Saint Petersburg, Russia
| |
Collapse
|
142
|
Abdel-Maksoud MS, Eitah HE, Hassan RM, Abd-Allah WH. Design and synthesis of novel pyrimidine-pyrazole hybrids with dual anticancer and anti-inflammatory effects targeting BRAFV600E and JNK. Mol Divers 2025:10.1007/s11030-025-11121-w. [PMID: 39985727 DOI: 10.1007/s11030-025-11121-w] [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: 10/29/2024] [Accepted: 01/18/2025] [Indexed: 02/24/2025]
Abstract
Two new series of pyrimidinyl ethyl pyrazoles derivatives 13a-f and 14a-f were designed and synthesized to possess both anticancer effect by inhibiting BRAFV600E and anti-inflammatory effect by inhibiting JNK isoforms. The structure of the new compounds was generated from hybridization of two main moieties. The pyrimidinyl moiety from reported BRAFV600E inhibitors, and the pyrazole moiety from JNK isoforms inhibitors. The new final compounds were tested on BRAFV600E, JNK1, JNK2, and JNK3 to measure their kinases inhibitory effect. Compound 14c showed the highest activity on JNK isoforms and BRAFV600E with IC50 = 0.51 μM, 0.53 μM, 1.02 μM, 0.009 μM on JNK1, JNK2, JNK3,and BRAFV600E, respectively. All final compounds were tested over four cancer cell lines related to the target enzymes. Compound 14d showed the most potent activity on all tested cell lines with IC50 = 0.87 μM, 0.91, 0.42 μM and 0.63 μM on MOLT-4, K-562, SK-MEL-28, and A375 cell lines, respectively. The ability of 14d and 14c to inhibit MEK1/2 and ERK1/2 phosphorylation was performed by using western blot. The cell cycle analysis of compound 14d on A375 cell line revealed that compound 14d arrested cell growth at G0-G1 phase. Compound 14d remarkably decreased cell migration compared to control group in traditional migration test. Compounds 13a-f and 14a-f showed significant ability to inhibit nitric oxide release and PGE2 production on raw 264.7 macrophages. Compounds 13d and 14d exhibited high inhibitory effect on iNOS and COX-2 compared to COX-1. Finally, the effect of most potent compounds on TNF-alpha and IL-6 was determined.
Collapse
Affiliation(s)
- Mohammed S Abdel-Maksoud
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), (ID: 60014618), Dokki, P.O. 12622, Giza, Egypt.
| | - Hebatollah E Eitah
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC) (Pharmacology Group), (ID: 60014618), Dokki, P.O. 12622, Giza, Egypt
| | - Rasha M Hassan
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), (ID: 60014618), Dokki, P.O. 12622, Giza, Egypt
| | - Walaa Hamada Abd-Allah
- Pharmaceutical Chemistry Department, Collage of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, P.O. 77, 6th of October City, Giza, Egypt
| |
Collapse
|
143
|
Sukumaran RA, Lakavath K, Phani Kumar VVN, Karingula S, Mahato K, Kotagiri YG. Eco-friendly synthesis of a porous reduced graphene oxide-polypyrrole-gold nanoparticle hybrid nanocomposite for electrochemical detection of methotrexate using a strip sensor. NANOSCALE 2025; 17:4472-4484. [PMID: 39807059 DOI: 10.1039/d4nr04010d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Chemotherapy is a crucial cancer treatment, but its effectiveness requires precise monitoring of drug concentrations in patients. This study introduces an innovative electrochemical strip sensor design to detect and continuously monitor methotrexate (MTX), a key chemotherapeutic drug. The sensor is crafted through an eco-friendly synthesis process that produces porous reduced graphene oxide (PrGO), which is then integrated with gold nanocomposites and polypyrrole (PPy) to boost the performance of a screen-printed carbon electrode (SPCE). Advanced techniques were employed for detailed characterization of the nanocomposites such as X-ray diffraction (XRD), Raman spectroscopy, X-ray photoelectron spectroscopy (XPS), field emission scanning electron microscopy (FE-SEM), and BET analysis. The enhanced sensor exhibited a notable increase in the electrochemical oxidation signals of MTX, attributed to the improved electron transfer at the SPCE/PrGO-PPy-Au electrode interface. Superior electrochemical interfacial properties were well characterized with the techniques of cyclic voltammetry, electrochemical impedance spectroscopy, and square wave voltammetry. The sensor demonstrates an efficient electrochemical response toward the detection of MTX with a broad detection range from 130 nM to 1 μM, an impressively low detection limit of 0.4 nM in human serum, and a sensitivity of 24.1 μA μM-1. This combination highlights its exceptional performance in detecting analytes with high precision and sensitivity. The sensor exhibited a long-term continuous monitoring stability response to monitor the MTX drug in human serum for 4 hours. The sensor's high sensitivity, selectivity, reproducibility, and stability over time emphasize its potential as a valuable tool for the swift on-site testing of anticancer drugs in clinical and environmental settings.
Collapse
Affiliation(s)
- Reshmi A Sukumaran
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678 557, India.
| | - Kavitha Lakavath
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678 557, India.
| | - V V N Phani Kumar
- Centre for Automotive Energy Materials, International Advanced Research Centre for Powder Metallurgy and New Materials (ARCI), Chennai 600113, Tamil Nadu, India
| | - Sampath Karingula
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678 557, India.
| | - Kuldeep Mahato
- Department of Nanoengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Yugender Goud Kotagiri
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala 678 557, India.
| |
Collapse
|
144
|
Alosaimi EH. Recent Developments in Colorimetric and Fluorimetric Chemosensors for the Detection of Mn 2+ Ions: A Review (2010-2024). Crit Rev Anal Chem 2025:1-21. [PMID: 39969414 DOI: 10.1080/10408347.2025.2460091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Manganese is an essential metal ion involved in various biological and environmental processes, but its excess can lead to toxicity, particularly affecting the nervous system. Therefore, developing selective and sensitive detection methods for Mn2+ ions is of paramount importance. Colorimetric and fluorimetric chemosensors have emerged as promising tools for the detection of Mn2+ due to their simplicity, cost-effectiveness, and real-time monitoring capabilities. This review discusses recent advances in the colorimetric and fluorimetric chemosensors that exhibit distinct color or fluorescence changes upon interaction with Mn2+ ions. The review explores different organic and nanomaterials, focusing on their mechanisms of sensing, sensitivity, selectivity, and practical applications in environmental monitoring, healthcare, and food safety. The article also provides insights into future research directions aimed at overcoming these challenges, improving chemosensor performance, and expanding the applicability of colorimetric and fluorimetric chemosensors for Mn2+ detection in diverse real-world scenarios.
Collapse
Affiliation(s)
- Eid H Alosaimi
- Department of Chemistry, College of Science, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
145
|
Alqarni SS, Khan NU. Integrating alternative therapies in overcoming chemotherapy resistance in tumors. Mol Biol Rep 2025; 52:239. [PMID: 39961936 DOI: 10.1007/s11033-025-10361-1] [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: 10/08/2024] [Accepted: 02/11/2025] [Indexed: 05/09/2025]
Abstract
Chemotherapy-resistant tumors present a significant challenge in oncology, often leading to treatment failures owing to mechanisms such as genetic mutations, drug efflux, altered metabolism, and adaptations within the tumor microenvironment. These factors limit the effectiveness of treatment and contribute to tumor resistance. This review highlights the role of alternative therapies aimed at overcoming resistance mechanisms. Several alternative strategies with high efficacy rate against tumor resistance are being explored, including targeted therapies (58-64%), immunotherapy (80%), hormone therapy (22-61%), and emerging approaches such as herbal therapies (90%), probiotics (34-90%), metabolic therapies (> 50%), epigenetic therapies (51-89%), microbiome-based therapies (50%), gene therapy (67-80%), photodynamic therapy/hypothermia (86-99%), and nanotechnology (50-67%). Integrating these alternative strategies with conventional treatments has the potent-al to augment the therapeutic efficacy and patient outcomes. Despite this progress, limitations in cancer therapeutics include the lack of predictive biomarkers, resistance mechanisms, and tumor heterogeneity, all of which contribute to treatment failure and relapse. To address these limitations, advancements in molecular diagnostics, as well as early detection through liquid biopsies, and the use of biomarkers to monitor resistance and guide treatment are crucial. Additionally, expanding clinical trials is essential to validate new therapies and improve patient outcomes.
Collapse
Affiliation(s)
- Sana S Alqarni
- Department of Clinical Laboratory Science, College of Applied Medical Science, King Saud University, 11421, Riyadh, Saudi Arabia
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, 25130, Pakistan.
| |
Collapse
|
146
|
Mahnoor, Malik K, Kazmi A, Sultana T, Raja NI, Bibi Y, Abbas M, Badruddin IA, Ali MM, Bashir MN. A mechanistic overview on green assisted formulation of nanocomposites and their multifunctional role in biomedical applications. Heliyon 2025; 11:e41654. [PMID: 39916856 PMCID: PMC11800088 DOI: 10.1016/j.heliyon.2025.e41654] [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: 06/12/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
The importance of nanocomposites constantly attains attention because of their unique properties all across the fields especially in medical perspectives. The study of green-synthesized nanocomposites has grown to be extremely fascinating in the field of research. Nanocomposites are more promising than mono-metallic nanoparticles because they exhibit synergistic effects. This review encapsulates the current development in the formulation of plant-mediated nanocomposites by using several plant species and the impact of secondary metabolites on their biocompatible functioning. Phyto-synthesis produces diverse nanomaterials with biocompatibility, environment-friendliness, and in vivo actions, characterized by varying sizes, shapes, and biochemical nature. This process is advantageous to conventional physical and chemical procedures. New studies have been conducted to determine the biomedical efficacy of nanocomposites against various diseases. Unfortunately, there has been inadequate investigation into green-assisted nanocomposites. Incorporating phytosynthesized nanocomposites in therapeutic interventions not only enhances healing processes but also augments the host's immune defenses against infections. This review highlights the phytosynthesis of nanocomposites and their various biomedical applications, including antibacterial, antidiabetic, antiviral, antioxidant, antifungal, anti-cancer, and other applications, as well as their toxicity. This review also explores the mechanistic action of nanocomposites to achieve their designated tasks. Biogenic nanocomposites for multimodal imaging have the potential to exchange the conventional methods and materials in biomedical research. Well-designed nanocomposites have the potential to be utilized in various biomedical fields as innovative theranostic agents with the subsequent objective of efficiently diagnosing and treating a variety of human disorders.
Collapse
Affiliation(s)
- Mahnoor
- Department of Botany, PMAS, Arid Agriculture University Rawalpindi, Pakistan
| | - Khafsa Malik
- Department of Botany, PMAS, Arid Agriculture University Rawalpindi, Pakistan
| | - Abeer Kazmi
- The State Key Laboratory of Freshwater Ecology and Biotechnology, The Key Laboratory of Aquatic Biodiversity and Conservation of Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, Hubei, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Tahira Sultana
- Department of Botany, PMAS, Arid Agriculture University Rawalpindi, Pakistan
| | - Naveed Iqbal Raja
- Department of Botany, PMAS, Arid Agriculture University Rawalpindi, Pakistan
| | - Yamin Bibi
- Department of Botany, Rawalpindi Women University, Rawalpindi, Pakistan
| | - Mazhar Abbas
- Department of Biochemistry, University of Veterinary and Animal Science Lahore (Jhang Campus), Jhang, 35200, Pakistan
| | - Irfan Anjum Badruddin
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - M. Mahmood Ali
- Department of Mechatronic Engineering, Atlantic Technological University Sligo, Ash Lane, F91 YW50, Sligo, Ireland
| | - Muhammad Nasir Bashir
- Department of Mechanical Engineering, Yonsei University, Seoul, 120-749, Republic of Korea
- National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
147
|
Sharma S, Kaur V, Duhan P, Singh R, Agnihotri N. Evaluation of Anticancer Activity of Novel and Tumor-Targeted Glutamine-Conjugated Organotin(IV) Compounds in Colorectal Cancer─An In Vitro and In Vivo Study. J Med Chem 2025; 68:2593-2607. [PMID: 39834112 DOI: 10.1021/acs.jmedchem.4c01728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Over the years, numerous ligand-based organotin(IV) Schiff base compounds have shown remarkable cytotoxicity and anticancer activities, but their clinical use is restricted by systemic toxicity, prompting the search for targeted therapies. Targeted delivery can be enhanced by exploiting the inherent characteristics of cancer cells such as glutamine addiction, which is essential to support cellular biosynthesis and cell growth to sustain aberrant proliferation. Our previous study revealed glutamine-conjugated organotin(IV) compounds have strong DNA/protein affinities, favorable in silico ADME profiles, and significant antiproliferative activity. In this study, these compounds demonstrated significant cytotoxicity against human colon carcinoma and adenocarcinoma cell lines via the induction of cell cycle arrest and apoptosis. In DMH/DSS-induced experimental colon carcinogenesis, these compounds reduced tumor burden and volume and inhibited cell proliferation and induced apoptosis, with minimal toxicity. Tissue distribution studies revealed selective accumulation in the colon. These findings support their potential as chemotherapeutic candidates for colon cancer.
Collapse
Affiliation(s)
- Shagun Sharma
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Varinder Kaur
- Department of Chemistry, Panjab University, Chandigarh 160014, India
| | - Pratibha Duhan
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| | - Raghubir Singh
- Department of Chemistry, DAV College, Sector 10, Chandigarh 160011, India
| | - Navneet Agnihotri
- Department of Biochemistry, Panjab University, Chandigarh 160014, India
| |
Collapse
|
148
|
Trybus W, Trybus E, Obarzanowski M, Król T. Quinalizarin induces autophagy, apoptosis and mitotic catastrophe in cervical and prostate cancer cells. Sci Rep 2025; 15:5252. [PMID: 39939343 PMCID: PMC11822151 DOI: 10.1038/s41598-025-89847-8] [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: 11/20/2024] [Accepted: 02/07/2025] [Indexed: 02/14/2025] Open
Abstract
Cancer diseases are a serious health problem for society, and among them cervical and prostate cancer rank high in terms of mortality. One of the reasons is the phenomenon of drug resistance and side effects accompanying conventional chemo- and radiotherapy. This requires continuous development of alternative treatment methods and searching for new compounds with anti-cancer potential. An example is quinalizarin, which was tested for its anti-cancer potential. The MTT test showed cytotoxic activity of quinalizarin against Hela and DU145 cell lines. Morphological analysis showed nuclear changes typical of apoptosis, which was confirmed by the annexin V/PE test, activation of caspases 3/7 and inhibition of Bcl-2 protein expression. Increased permeability of mitochondrial membranes and ROS generation were demonstrated. Inhibition of cell migration, blocking in the G0/G1 phase, increased number of cells with damaged DNA and an increase in markers of mitotic catastrophe, i.e. micro- and multinucleation including the presence of abnormal mitotic figures were also observed. At the same time, increased autophagy was observed, and preincubation of cells with chloroquine inhibited this process, which contributed to the increased cytotoxicity of quinalizarin towards the tested cells. Quinalizarin has a multidirectional effect based on apoptosis and alternative types of cell death.
Collapse
Affiliation(s)
- Wojciech Trybus
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland.
| | - Ewa Trybus
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland.
| | - Mateusz Obarzanowski
- Department of Oncology, Medical College, Jan Kochanowski University of Kielce, al. IX Wieków Kielc 19a, Kielce, 25-516, Poland
- Department of Urology, Holy Cross Cancer Center, Stefana Artwińskiego, Kielce, 25-734, Poland
| | - Teodora Król
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland
| |
Collapse
|
149
|
Noh I, Guo Z, Wang R, Zhu AT, Krishnan N, Mohapatra A, Gao W, Fang RH, Zhang L. Modular functionalization of cellular nanodiscs enables targeted delivery of chemotherapeutics into tumors. J Control Release 2025; 378:145-152. [PMID: 39657891 PMCID: PMC11933242 DOI: 10.1016/j.jconrel.2024.12.004] [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: 07/22/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
The effective delivery of chemotherapeutic drugs to tumor sites is critical for cancer treatment and remains a significant challenge. The advent of nanomedicine has provided additional avenues for altering the in vivo distribution of drug payloads and increasing tumor localization. More recently, cell-derived nanoparticles, with their biocompatibility and unique biointerfacing properties, have demonstrated considerable utility for drug delivery applications. Here, we demonstrate that cell membrane-derived nanodiscs can be employed for tumor-targeted delivery. To bestow active targeting capabilities to the cellular nanodiscs, we utilize a modular functionalization strategy based on the SpyCatcher system. This enables the nanodiscs to be covalently modified with any targeting ligand labeled with a short SpyTag peptide sequence. As a proof-of-concept, a model chemotherapeutic doxorubicin is loaded into nanodiscs functionalized with an affibody targeting epidermal growth factor receptor. The resulting nanoformulation demonstrates strong tumor targeting both in vitro and in vivo, and it is able to significantly inhibit tumor growth in a murine breast cancer model.
Collapse
Affiliation(s)
- Ilkoo Noh
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhongyuan Guo
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Rui Wang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Audrey T Zhu
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Nishta Krishnan
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Animesh Mohapatra
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA; Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
150
|
Murg SI, Matiș L, Moldovan AF, Popovici DI, Negru AG, Ghitea TC, Popescu MI. Association Between Advanced TNM Stages and Increased Risk of Cardiac Dysfunction in Patients with LVEF < 50. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:301. [PMID: 40005419 PMCID: PMC11857368 DOI: 10.3390/medicina61020301] [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] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/30/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Cardio-oncology addresses the growing concern of cardiovascular complications arising from cancer therapies. Although cancer treatments have greatly enhanced survival outcomes, they frequently carry substantial risks to cardiovascular health. This research examines the cardiovascular toxicity associated with HER2-targeted therapies, focusing on the interconnection between tumor characteristics, including histopathological profiles and TNM classification, and the development of cardiovascular complications. The objective is to identify key correlations that inform better prevention and management strategies for cardiotoxicity in oncology patients. Materials and Methods: This retrospective study analyzed cancer patients undergoing cytostatic treatments, particularly anthracyclines, radiotherapy, and HER2-targeted therapies. Cardiac function was monitored using echocardiographic assessments, including global longitudinal strain and left ventricular ejection fraction (LVEF). Patients were stratified based on TNM cancer staging and histopathological findings to evaluate correlations between treatment regimens and cardiovascular outcomes. Results: The analysis revealed a significant association between advanced TNM stages and reduced LVEF, with patients in stage T4 showing the highest prevalence of cardiac dysfunction. Cytostatic treatments, such as anthracyclines and HER2-targeted therapies, were identified as key contributors to cardiotoxicity, particularly in advanced-stage cancer patients. These findings emphasize the importance of regular cardiac monitoring to detect early signs of cardiotoxicity, as patients with pre-existing cardiovascular risk factors demonstrated a higher prevalence of complications. Conclusions: This study highlights the need for personalized treatment approaches and tailored cardioprotective strategies to improve outcomes and enhance the quality of life for oncology patients. Future studies should prioritize developing improved strategies to reduce the cardiovascular complications linked to contemporary cancer treatments.
Collapse
Affiliation(s)
- Sergiu Ioan Murg
- Doctoral School, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania;
| | - Loredana Matiș
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Andrada Florina Moldovan
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Dorel Ionel Popovici
- Department of Oncology, Faculty of Medicine, Victor Babeş University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Alina Gabriela Negru
- Department of Cardiovascular Diseases of Timisoara, Victor Babeş University of Medicine and Pharmacy Timisoara, G. Adam Str. No. 13A, 300310 Timisoara, Romania;
| | - Timea Claudia Ghitea
- Pharmacy Department, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania
| | - Mircea Ioachim Popescu
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| |
Collapse
|