1
|
Roman A, Motoc A, Marcovici I, Dehelean C, Nicolescu L, Boru C. Genistein Improves the Cytotoxic, Apoptotic, and Oxidative-Stress-Inducing Properties of Doxorubicin in SK-MEL-28 Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:798. [PMID: 40428756 PMCID: PMC12113575 DOI: 10.3390/medicina61050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Cutaneous melanoma (CM) poses a continuous challenge in oncology due to the developing resistance to available treatments. Doxorubicin (DOX) is noted as one of the most effective chemotherapeutics, although associated toxicity and resistance limit its use in CM treatment. Consequently, DOX has become a promising candidate for combination therapies targeting this neoplasm. Genistein (GEN) gathered significant attention due to its anti-neoplastic properties and ability to enhance the effects of DOX against several cancers, yet this association remains underexplored in CM. Therefore, this study investigated the combination therapy regimen comprising GEN and DOX in terms of anti-melanoma activity and safety profile. Materials and Methods: The in vitro experiments were performed on SK-MEL-28 and HaCaT cells. Cell viability was determined using MTT assay. Cell morphology and confluence were inspected microscopically. Nuclear and cytoskeletal aspects were assessed via immunofluorescence. Apoptosis and oxidative stress were quantified through caspase activity and intracellular reactive oxygen species (ROS) production, respectively. The irritant effect was evaluated on the chorioallantoic membrane. Results: The results revealed that the combination of GEN 10 µM with DOX (0.5 and 1 µM) provided augmented cytotoxic events (e.g., reduced cell viability, altered cell morphology and confluence, apoptotic-like impairments in nuclear shape and cytoskeletal network, increased caspases-3/7 and -9 activity, and elevated ROS) in SK-MEL-28 cells, compared to individual treatments, and exerted a strong synergistic interaction. Simultaneously, GEN 10 µM efficiently surpassed the toxic effects (e.g., viability and confluence loss, hypertrophy, and cytoskeletal condensation) of DOX (0.5 and 1 µM) in HaCaT cells. In ovo, GEN 10 µM + DOX 1 µM treatment was classified as non-irritant. Conclusions: These findings stand as one of the first contributions revealing the beneficial therapeutic interplay between GEN and DOX at physiologically achievable concentrations that resulted in elevated anti-tumor properties in CM cells and alleviated toxicity in keratinocytes.
Collapse
Affiliation(s)
- Andrea Roman
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Andrei Motoc
- Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Cristina Dehelean
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Laura Nicolescu
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
| | - Casiana Boru
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, 94 Revolutiei Blvd., 310130 Arad, Romania
| |
Collapse
|
2
|
Liu J, Fu W, Wang X, Liang Z, Meng F. The role of HDAC2 inhibition in cardioprotection against doxorubicin-induced myocardial injury. Front Cardiovasc Med 2025; 12:1557119. [PMID: 40078459 PMCID: PMC11897267 DOI: 10.3389/fcvm.2025.1557119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction The molecular mechanisms underlying cardioprotection against doxorubicin (DOX)-induced myocardial injury are poorly understood. Histone deacetylase 2 (HDAC2) plays a significant role in oxidative stress, apoptosis, and mitochondrial dysfunction and is implicated in many human diseases, This study investigated the relationship between HDAC2 expression and DOX-induced myocardial injury using the in vivo rat model of DOX-induced cardiotoxicity and in vitro experiments with the H9c2 cardiomyocytes. Methods The rat model of DOX-induced myocardial injury was established by administering DOX via intraperitoneal injections. HDAC2 expression was suppressed by administering rats with sodium butyrate (SB) via intraperitoneal injections. Echocardiography measurements were performed at baseline and on day 15 post-treatment. The rats were euthanized on day 15 and cardiac tissues were harvested. The cardiac tissue samples were analyzed by hematoxylin and eosin H&E staining, immunohistochemistry, Masson staining, Sirius Red staining, TUNEL staining, and western blotting to determine the status of HDAC2 expression and myocardial apoptosis. In the vitro experiments, H9c2 cells were treated with DOX. HDAC2 expression was suppressed using sodium butyrate or transfected cells with the shRNA knockdown HDAC2 (shHDAC2). The H9c2 cells from different groups were analyzed by Rt-qPCR, CCK-8 cell viability assay, and western blotting to determine the status of HDAC2 expression and cardiomyocyte apoptosis. Results DOX treatment induced cardiac dysfunction in rats. The cardiac tissues of the DOX-treated rats and H9c2 cells showed significantly higher levels of HDAC2 compared to the corresponding controls. However, inhibition of HDAC2 significantly mitigated DOX-induced myocardial injury in rats. This suggested a strong association between HDAC2 expression and DOX-induced myocardial injury. In the H9c2 cells, HDAC2 knockdown by shHDAC2 alleviated DOX-induced apoptosis by enhacing AKT phosphorylation. These findings demonstrated that HDAC2 silencing protected against DOX-induced cardiomyocyte apoptosis by activating the PI3K/AKT signaling pathway. Conclusion Suppressing HDAC2 protected against DOX-induced cardiomyocyte apoptosis by activating the PI3K/AKT signaling pathway. Therefore, HDAC2 is a promising therapeutic target for mitigating DOX-induced myocardial injury.
Collapse
Affiliation(s)
- Jinsha Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Center, Changchun, China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xue Wang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Center, Changchun, China
| | - Zuowen Liang
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Fanbo Meng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
- Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Cardiovascular Research Center, Changchun, China
| |
Collapse
|
3
|
Kodel HDAC, Alizadeh P, Ebrahimi SN, Machado TOX, Oliveira MBPP, Fathi F, Souto EB. Liposomes and Niosomes: New trends and applications in the delivery of bioactive agents for cancer therapy. Int J Pharm 2025; 668:124994. [PMID: 39586512 DOI: 10.1016/j.ijpharm.2024.124994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lipid-based nanocarriers have been in continuous development as strategies to enhance drug delivery efficiency. Liposomes are delivery systems primarily composed of phospholipids and cholesterol (or other suitable stabilizers) that have transformed the pharmaceutical field by improving drug targeting and release control. The success of this technology is strongly attributed to phospholipids, which are components of cell membranes, forming a biocompatible system. Nevertheless, drawbacks related to their production cost and stability under certain conditions led to the development of niosomes by replacing phospholipids with non-ionic surfactants. Both liposomes and niosomes have been widely studied and optimized for the delivery of bioactive agents targeting many diseases, including cancer. They can improve the efficacy of cancer therapy by reducing toxicity and off-target effects. Due to the complexity of this disease, many approaches should be considered, and the composition and physical properties of liposomes and niosomes influence the outcomes. In this review, we discuss the role of liposomes and niosomes in delivering bioactives for cancer therapy, emphasizing their specific characteristics, associated challenges, and the latest advancements aimed at enhancing their effectiveness.
Collapse
Affiliation(s)
- Helena de A C Kodel
- Rede Nordeste de Biotecnologia-RENORBIO, University of Tiradentes, Farolândia, 49010-390, Aracaju, Sergipe, Brazil
| | - Paria Alizadeh
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Samad N Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Tatiane O X Machado
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy of University of Porto, Jorge de Viterbo Ferreira, 4050-313, Porto, Portugal; Department of Agroindustry, Federal Institute of Sertão Pernambucano, Campus Petrolina Zona Rural, PE 647, Km 22, PISNC N4, 56302-970, Petrolina, Pernambuco, Brazil
| | - M Beatriz P P Oliveira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Faezeh Fathi
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Eliana B Souto
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
4
|
Mohareb RM, Abdelaziz MA, Jame R, Omer N, Labib HM. Synthesis of New Chromen-5-one Derivatives from Dimedone and their Antiproliferative Evaluations against Selected Cancer Cell Lines Together with Hepatocellular Carcinoma and Cervical Carcinoma. Anticancer Agents Med Chem 2025; 25:134-149. [PMID: 39301897 DOI: 10.2174/0118715206323592240909101754] [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: 05/05/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The coumarin nuclei, which exist in many heterocyclic compounds, has gained a lot of attention over the past decade due to their wide range of biological activities such as antibacterial, anticoagulant, antiviral, antifungal, anticancer, and anti-inflammatory properties. OBJECTIVE The multi-component reactions of 5,5-dimethylcyclohexane-1,3-dione with acetophenone derivatives and triethoxymethane produced biologically active target chromene molecules and their fused derivatives. METHODS The reaction of 5,5-dimethylcyclohexane-1,3-dione and each of triethoxymethane and acetophenone derivatives 3a-g in absolute ethanol containing triethylamine gave the 4,6,7,8-tetrahydro-5H-chromen-5-one derivatives 4a-g. Compounds 4a-d were used for further heterocyclization reactions to produce biologically active fused pyrazole, thiophene, and thiazole derivative corporate with the chromenes caffold. RESULTS The cytotoxicity of the synthesized compounds were evaluated using six cancer cell lines together with c-Met kinase and PC-3 cell line inhibitions. In addition, cytotoxicity toward hepatocellular carcinoma HepG2 and cervical carcinoma HeLa was carried out as well as the in-vitro cytotoxic potential for all compounds against peripheral blood lymphocytes (PBL) extracted from healthy donors. Morphological changes of the A549 cell line by the two most active compounds were also studied. CONCLUSION The synthesized heterocyclic compounds were originally obtained from 5,5-dimethylcyclohexane- 1,3-dione. Several of the produced compounds exhibited high inhibitions toward several cancer cell lines proving high inhibitions, therefore, encouraging further studies to synthesize heterocyclic compounds based on chromene scaffold.
Collapse
Affiliation(s)
- Rafat Milad Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, A. R. Egypt
| | - Mahmoud A Abdelaziz
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Kingdom of Saudi Arabia
| | - Rasha Jame
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Kingdom of Saudi Arabia
| | - Noha Omer
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Kingdom of Saudi Arabia
| | - Hanan Maged Labib
- Department of chemistry, Faculty of Science, Cairo University, Giza, A. R. Egypt
| |
Collapse
|
5
|
Schiavoni V, Emanuelli M, Sartini D, Salvolini E, Pozzi V, Campagna R. Curcumin and its Analogues in Oral Squamous Cell Carcinoma: State-of-the-art and Therapeutic Potential. Anticancer Agents Med Chem 2025; 25:313-329. [PMID: 38757321 DOI: 10.2174/0118715206297840240510063330] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 05/18/2024]
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common cancer arising from squamous epithelium in the oral cavity and is characterized by high aggressiveness and metastatic potential, which together with a late diagnosis results in a 5-year survival rate of only 50% of patients. The therapeutic options for OSCC management are limited and largely influenced by the cancer stage. While radical surgery can be curative in early stage of disease, most cases require adjuvant therapies, including chemotherapy and radiotherapy which, however, often achieve poor curative rates and are associated with important negative effects. Therefore, there is an urgent need to discover new alternative treatment strategies to improve patients' outcomes. Several medicinal herbs are being studied for their preventive or therapeutic effect in several diseases, including cancer. In particular, the Indian spice curcumin, largely used in oriental countries, has been studied as a chemopreventive or adjuvant agent for different malignancies. Indeed, curcumin is characterized by important biological properties, including antioxidant, anti-inflammatory, and anticancer effects, which could also be exploited in OSCC. However, due to its limited bioavailability and poor aqueous solubility, this review is focused on studies designing new synthetic analogues and developing novel types of curcumin delivery systems to improve its pharmacokinetic and biological properties. Thus, this review analyses the potential therapeutic role of curcumin in OSCC by providing an overview of current in vitro and in vivo studies demonstrating the beneficial effects of curcumin and its analogues in OSCC.
Collapse
Affiliation(s)
- Valentina Schiavoni
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, 60131, Italy
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| |
Collapse
|
6
|
Taghikhani A, Babazadeh M, Davaran S, Ghasemi E. Facile preparation of a pH-sensitive biocompatible nanocarrier based on magnetic layered double hydroxides/Cu MOFs-chitosan crosslinked к-carrageenan for controlled doxorubicin delivery to breast cancer cells. Colloids Surf B Biointerfaces 2024; 243:114122. [PMID: 39079184 DOI: 10.1016/j.colsurfb.2024.114122] [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: 04/29/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 09/17/2024]
Abstract
Recently, the biocompatibility of hydrogel nanoparticles has gained considerable research attention in the field of drug delivery. In this regard, we design a pH-controlled nanocarrier based on magnetic layered double hydroxides/copper metal-organic framework-chitosan crosslinked к-carrageenan hydrogel nanoparticles (LDH-Fe3O4/Cu MOF-DOX-CS@CAR) for targeted release from DOX to breast cancer cells. FT-IR, EDX, XRD, FE-SEM, VSM, and Zeta potential investigated the chemical structure of hydrogel nanoparticles. The encapsulation efficiency and drug loading capacity of the DOX were obtained to be 96.1 % and 9.6 %, respectively. The cumulative release of DOX from LDH-Fe3O4/Cu MOF-DOX-CS@CAR at pH 5.5 and 7.4 after 72 h was 60.3 % and 22.6 %, respectively. These in vitro release results confirmed the controlled release and pH-response behavior of hydrogel nanoparticles. Also, the mechanism of DOX release from LDH-Fe3O4/Cu MOF-DOX-CS@CAR hydrogel nanoparticles showed that the Korsmeyer-Peppas model with Fickian diffusion is the best-fitting model for describing the release behavior of DOX from hydrogel nanoparticles. The cellular cytotoxicity and DAPI tests of the prepared LDH and LDH-Fe3O4/Cu MOF toward L929 non-cancerous cells and MCF-7 breast cancer cells confirm its relative biocompatibility and safety. Whereas, LDH-Fe3O4/Cu MOF-DOX-CS@CAR hydrogel nanoparticles toward MCF-7 breast cancer cells had higher cytotoxicity effects due to the targeted and controlled release of DOX to MCF-7 cells. The in vitro DPPH, hemolysis assay, colloidal stability, and enzymatic degradation proved the excellent antioxidant activity (71.81 %), blood compatibility (less than 5 %), better stability, and biodegradation behavior of hydrogel nanoparticles. On these findings, the present study suggests the potential of the prepared LDH-Fe3O4/Cu MOF-DOX-CS@CAR hydrogel nanoparticles as a pH-controlled drug delivery system for cancer treatment and various biomedical uses.
Collapse
Affiliation(s)
- Afsaneh Taghikhani
- Department of Chemistry, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mirzaagha Babazadeh
- Department of Chemistry, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | - Soodabeh Davaran
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Ghasemi
- Department of Chemistry, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
7
|
Nalinbenjapun S, Sripetthong S, Basit A, Suksuwan A, Sajomsang W, Ovatlarnporn C. Fabrication of curcumin-loaded nano-micelles based on quercetin-quarternary ammonium-chitosan (Qu-QCS) conjugate and evaluation of synergistic effect with doxorubicin against breast cancer. Int J Biol Macromol 2024; 281:135904. [PMID: 39482127 DOI: 10.1016/j.ijbiomac.2024.135904] [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: 04/07/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 11/03/2024]
Abstract
The applications of quarternized chitosans have achieved notable success in the development of drug-delivery systems. This study reported the preparation of quercetin-quarternized chitosan (Qu-QCS) conjugate and its application for the fabrication of stable and safe curcumin (cur) loaded nano-micelles with high targeting ability and selectivity towards the breast cancer cell lines. Moreover, doxorubicin (dox) was co-treated with the nanomicelles to enhance the efficacy and reduce the cardiotoxic effects of dox. Structural properties of Qu-QCS were evaluated by FTIR, DSC, and XRD analysis and the yield obtained was 48.82 %. The nano-micelles obtained showed spherical shape, <200 nm size, 48.38 % entrapment efficiency, prolonged stability at 4 °C and pH-responsive release pattern. The cur-loaded nano-micelles showed higher activity and selectivity against breast cancer (MCF-7 and MDA-MB-231) cell lines with enhanced internalization, lower toxicity to the normal cardiomyoctyes (H9C2), enhanced the cell cycle arrest at the G2/M phase of the breast cancer cell lines and induced apoptosis with high intensity compared to pure cur. Moreover, the co-treatment of dox with cur-loaded Qu-QCS nano-micelles showed increased anticancer activity and reduced cardiotoxicity. Overall, this study suggests the potential applications of cur-loaded Qu-QCS micelles in the delivery of chemotherapeutic agents and complementary support in combination with chemotherapeutic agents.
Collapse
Affiliation(s)
- Sirinporn Nalinbenjapun
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Sasikarn Sripetthong
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Abdul Basit
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand.
| | - Acharee Suksuwan
- The Halal Science Center, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warayuth Sajomsang
- Nanodelivery System Laboratory, National Nanotechnology Center, National Science and Technology Development Agency, Phathum Thani 12120, Thailand
| | - Chitchamai Ovatlarnporn
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand.
| |
Collapse
|
8
|
Mayo B, Penroz S, Torres K, Simón L. Curcumin Administration Routes in Breast Cancer Treatment. Int J Mol Sci 2024; 25:11492. [PMID: 39519045 PMCID: PMC11546575 DOI: 10.3390/ijms252111492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Breast cancer is a public health concern worldwide, characterized by increasing incidence and mortality rates, requiring novel and effective therapeutic strategies. Curcumin is a bioactive compound extracted from turmeric with several pharmacological activities. Curcumin is a multifaceted anticancer agent through mechanisms including the modulation of signaling pathways, inhibition of cell proliferation, induction of apoptosis, and production of reactive oxygen species. However, the poor water solubility and bioavailability of curcumin create important barriers in its clinical application. This review elaborates on the therapeutic potential of curcumin in breast cancer treatment, focusing on the efficacy of different administration routes and synergistic effects with other therapeutic agents. The intravenous administration of curcumin-loaded nanoparticles significantly improves bioavailability and therapeutic outcomes compared to oral routes. Innovative formulations, such as nano-emulsifying drug delivery systems, have shown promise in enhancing oral bioavailability. While intravenous delivery ensures higher bioavailability and direct action on tumor cells, it is more invasive and expensive than oral administration. Advancing research on curcumin in breast cancer treatment is essential for improving therapeutic outcomes and enhancing the quality of life of patients.
Collapse
Affiliation(s)
| | | | - Keila Torres
- Nutrition and Dietetic School, Universidad Finis Terrae, Santiago 7501014, Chile; (B.M.); (S.P.)
| | - Layla Simón
- Nutrition and Dietetic School, Universidad Finis Terrae, Santiago 7501014, Chile; (B.M.); (S.P.)
| |
Collapse
|
9
|
Saharkhiz S, Nasri N, Naderi N, Dini G, Ghalehshahi SS, Firoozbakht F. Evaluating a targeted Palbociclib-Trastuzumab loaded smart niosome platform for treating HER2 positive breast cancer cells. Int J Pharm X 2024; 7:100237. [PMID: 38516198 PMCID: PMC10955288 DOI: 10.1016/j.ijpx.2024.100237] [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: 12/08/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
In this study, we present a targeted and pH-sensitive niosomal (pHSN) formulation, incorporating quantum dot (QD)-labeled Trastuzumab (Trz) molecules for the specific delivery of Palbociclib (Pal) to cells overexpressing human epidermal growth factor receptor 2 (HER2). FTIR analyses confirmed the successful preparation of the pHSNs and their bioconjugation. The labeled Trz-conjugated Pal-pHSNs (Trz-Pal-pHSNs) exhibited a size of approximately 170 nm, displaying a spherical shape with a neutral surface charge of -1.2 mV. Pal encapsulation reached ∼86%, and the release pattern followed a two-phase pH-dependent mechanism. MTT assessments demonstrated enhanced apoptosis induction, particularly in HER2-positive cells, by Trz-Pal-pHSNs. Fluorescence imaging further validated the internalization of particles into cells. In conclusion, Trz-Pal-pHSNs emerge as a promising platform for personalized medicine in the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Nazanin Naderi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Saeid Shirzadi Ghalehshahi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Fateme Firoozbakht
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| |
Collapse
|
10
|
Serini S, Calviello G. Potential of Natural Phenolic Compounds against Doxorubicin-Induced Chemobrain: Biological and Molecular Mechanisms Involved. Antioxidants (Basel) 2024; 13:486. [PMID: 38671933 PMCID: PMC11047710 DOI: 10.3390/antiox13040486] [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: 03/29/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Chemotherapy-induced cognitive impairment or "chemobrain" is a prevalent long-term complication of chemotherapy and one of the more devastating. Most of the studies performed so far to identify the cognitive dysfunctions induced by antineoplastic chemotherapies have been focused on treatment with anthracyclines, frequently administered to breast cancer patients, a population that, after treatment, shows a high possibility of long survival and, consequently, of chemobrain development. In the last few years, different possible strategies have been explored to prevent or reduce chemobrain induced by the anthracycline doxorubicin (DOX), known to promote oxidative stress and inflammation, which have been strongly implicated in the development of this brain dysfunction. Here, we have critically analyzed the results of the preclinical studies from the last few years that have evaluated the potential of phenolic compounds (PheCs), a large class of natural products able to exert powerful antioxidant and anti-inflammatory activities, in inhibiting DOX-induced chemobrain. Several PheCs belonging to different classes have been shown to be able to revert DOX-induced brain morphological damages and deficits associated with learning, memory, and exploratory behavior. We have analyzed the biological and molecular mechanisms implicated and suggested possible future perspectives in this research area.
Collapse
Affiliation(s)
- Simona Serini
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy
| | - Gabriella Calviello
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy
| |
Collapse
|
11
|
Alipournazari P, Pourmadadi M, Abdouss M, Rahdar A, Pandey S. Enhanced delivery of doxorubicin for breast cancer treatment using pH-sensitive starch/PVA/g-C 3N 4 hydrogel. Int J Biol Macromol 2024; 265:130901. [PMID: 38490383 DOI: 10.1016/j.ijbiomac.2024.130901] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
This study introduces a starch/PVA/g-C3N4 nanocarrier hydrogel for pH-sensitive DOX delivery in breast cancer. DOX was loaded into the nanocarrier with 44.75 % loading efficiency and 88 % Entrapment Efficiency. The release of DOX from the starch/PVA/g-C3N4 hydrogel was pH-sensitive: DOX was released faster in the acidic environment pertinent to cancer tumors (with a pH level of 5.4) than in the surrounding regular tissue environment carrying a more neutral environment (pH 7.4). The release kinetics analysis, encompassing zero-order, first-order, Higuchi, and Korsmeyer-Peppas models, revealed significant fitting with the Higuchi model at both pH 5.4 (R2 = 0.99, K = 9.89) and pH 7.4 (R2 = 0.99, K = 5.70) levels. Finally, we found that hydrogel was less damaging to healthy cells and more specific to apoptotic cells than the drug's free form. The starch/PVA/g-C3N4 hydrogel had low toxicity for both normal cells and breast cancer cells, whereas DOX loaded into the starch/PVA/g-C3N4 hydrogel had higher toxicity for cancer cells than the DOX-only control samples, and led to specific high apoptosis for cancer cells. The study suggests that DOX can be loaded into a starch/PVA/g-C3N4 hydrogel to improve the specificity of the drug's release in cancer tumors or in vitro breast cancer cells.
Collapse
Affiliation(s)
| | - Mehrab Pourmadadi
- Protein Research Center, Shahid Beheshti University, Tehran, GC 1983963113, Iran
| | - Majid Abdouss
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran.
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, Himachal Pradesh, India; Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
12
|
Ma B, Li Q, Mi Y, Zhang J, Tan W, Guo Z. pH-responsive nanogels with enhanced antioxidant and antitumor activities on drug delivery and smart drug release. Int J Biol Macromol 2024; 257:128590. [PMID: 38056756 DOI: 10.1016/j.ijbiomac.2023.128590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023]
Abstract
pH-responsive nanogels have played an increasingly momentous role in tumor treatment. The focus of this study is to design and develop pH-responsive benzimidazole-chitosan quaternary ammonium salt (BIMIXHAC) nanogels for the controlled release of doxorubicin hydrochloride (DOX) while enhancing its hydrophilicity. BIMIXHAC is crosslinked with carboxymethyl chitosan (CMC), hyaluronic acid sodium salt (HA), and sodium alginates (SA) using an ion crosslinking method. The chemical structure of chitosan derivatives was verified by 1H NMR and FT-IR techniques. Compared to hydroxypropyl trimethyl ammonium chloride chitosan (HACC)-based nanogels, BIMIXHAC-based nanogels exhibit better drug encapsulation efficiency and loading capacity (BIMIXHAC-D-HA 91.76 %, and 32.23 %), with pH-responsive release profiles and accelerated release in vitro. The series of nanogels formed by crosslinking with three different polyanionic crosslinkers have different particle size potentials and antioxidant properties. BIMIXHAC-HA, BIMIXHAC-SA and BIMIXHAC-CMC demonstrate favorable antioxidant capability. In addition, cytotoxicity tests showed that BIMIXHAC-based nanogels have high biocompatibility. BIMIXHAC-based nanogels exhibit preferable anticancer effects on MCF-7 and A549 cells. Furthermore, the BIMIXHAC-D-HA nanogel was 2.62 times less toxic than DOX to L929 cells. These results suggest that BIMIXHAC-based nanogels can serve as pH-responsive nanoplatforms for the delivery of anticancer drugs.
Collapse
Affiliation(s)
- Bing Ma
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Qing Li
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Yingqi Mi
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Jingjing Zhang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Wenqiang Tan
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; University of Chinese Academy of Sciences, Beijing 100049, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|