1
|
Kheraldine H, Hassan AF, Saeed S, Merhi M, Mateo JM, Ulamec M, Peric-Balja M, Vranic S, Al-Thawadi H, Moustafa AEA. Neratinib and metformin: A novel therapeutic approach against HER2-Positive Breast Cancer. Biomed Pharmacother 2025; 187:118034. [PMID: 40252335 DOI: 10.1016/j.biopha.2025.118034] [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: 02/12/2025] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND HER2-positive breast cancer (BC) is highly aggressive with a poor prognosis. It is driven by HER2 oncoprotein activation/crosstalk with other receptors like EGFR/(HER1), HER3, and HER4, in addition to IGF-1R, making these receptors ideal therapeutic targets as they are expressed/overexpressed in this subtype. We postulated that targeting HER2 and IGF-1R together is a promising therapy for HER2-positive BC. Thus, we explored the outcome of a novel combination treatment using neratinib, a pan-HER inhibitor, and metformin, an IGF-1R inhibitor, on HER2-positive BC cells. METHODS In this investigation, we used cellular and molecular biology techniques in addition to an angiogenesis model and tissue microarray analysis. RESULTS Our data revealed that this combination therapy significantly reduced cell viability compared to individual treatments and exhibited a synergistic effect in HER2-positive BC cells. Moreover, the combination disrupted cell cycle progression and inhibited colony formation, and invasion of HER2-positive BC cells; this is accompanied by the deregulation of HER1-3 and IGF-1R expression patterns, in addition to Caspase-3, BCL2, Fascin, and Vimentin. Moreover, key regulator molecular pathways, including, ERK1/2, AKT, p38 MAPK, and mTOR, were significantly downregulated upon treatment with neratinib and metformin combination. Additionally, our data pointed out that neratinib and metformin combination inhibited angiogenesis, in-ovo, an important biological event in cancer progression. Finally, using a cohort of 55 HER2-positive BC samples, we revealed that HER2 and IGF-1R are co-expressed in most of the cases. CONCLUSIONS These findings suggest that neratinib and metformin combination can present a promising strategy for targeting multiple pathways in HER2-positive BC.
Collapse
Affiliation(s)
- Hadeel Kheraldine
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Arij Fouzat Hassan
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Sumayyah Saeed
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Jericha Miles Mateo
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; Translational Cancer Research Facility, Interim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Monika Ulamec
- Clinical Department of Pathology and Cytology Ljudevit Jurak, Sister of Charity University Hospital Center, Zagreb, Croatia; Department of Pathology and Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Peric-Balja
- Oncological Pathology Department, Ljudevit Jurak Clinical Department of Pathology and Cytology, Sister of Charity University Hospital Center, Zagreb, Croatia
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hamda Al-Thawadi
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar; Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, Canada; ABS Research Review & Consultation, Montreal, QC, Canada.
| |
Collapse
|
2
|
Wu Y, Liu Y, Zhang Y, Wang X, Wang W. Targeted delivery of neratinib/xanthan gum-capped calcium carbonate nanoparticles induces apoptosis through PI3K/AKT pathway in breast cancer mice model. Int J Biol Macromol 2025; 310:142963. [PMID: 40222535 DOI: 10.1016/j.ijbiomac.2025.142963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Breast cancer (BC) continues to be the most common malignancy among women, presenting therapeutic challenges including drug resistance. This study examines the effectiveness of neratinib-loaded xanthan gum-capped calcium carbonate nanoparticles (NB/XG@CaCO₃NPs) for targeted breast cancer treatment. The nanoparticles were synthesized using the co-precipitation method, characterized, and assessed against MCF7 and MDA-MB231 breast cancer cell lines. In vitro, NB/XG@CaCO₃NPs demonstrated considerable cytotoxicity at approximately 50 μg/mL, whereas non-cancerous HMEC cells retained high viability. Flow cytometry demonstrated an 85.2 % apoptosis rate, signifying effective cancer cell mortality. Mechanistic investigations validated that the downregulation of the PI3K/AKT pathway facilitated the anti-tumor effects. In vivo, NB/XG@CaCO₃NPs administered intravenously to cadmium chloride-induced breast cancer mice significantly diminished tumor volume and enhanced histomorphology without causing major organ toxicity. qRT-PCR and western blot analysis further confirmed tumor suppression at the molecular level. These results indicate that NB/XG@CaCO₃NPs present a viable targeted treatment for BC, efficiently suppressing tumor proliferation while maintaining biocompatibility.
Collapse
Affiliation(s)
- Yilin Wu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yanxi Liu
- Department of Plastic and Reconstructive Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yawen Zhang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xuekui Wang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wan Wang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
3
|
Sharma G, Panwar R, Saini S, Tuli HS, Wadhwa K, Pahwa R. Emerging phytochemical-based nanocarriers: redefining the perspectives of breast cancer therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04003-3. [PMID: 40137964 DOI: 10.1007/s00210-025-04003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
Breast cancer is recognized as the most prevalent condition impacting women globally, despite several advancements in diagnosis and treatment. Existing therapeutic interventions including surgical procedures, radiation therapy, and chemotherapy often produce harmful effects on healthy tissues, trigger chemo-resistance, and augment the risk of relapse. In response to several unmet challenges, substantial research has been conducted to explore the therapeutic potential of natural compounds for breast cancer therapy. Progress in phytochemistry and pharmacology has facilitated the identification of diverse herbal bioactives with favorable safety profiles and multi-target mechanisms of action against breast cancer cells. Several phytochemicals like flavonoids and tannins have shown significant anticancer potential against breast cancer in diverse preclinical models. However, challenges like limited cellular absorption, low water solubility, and high molecular weight hinder their effective translation into clinical applications. Therefore, the development of novel therapies is imperative for overcoming these hurdles in breast cancer treatment effectively. Nanotechnology has reflected considerable perspective in tackling diverse challenges by encapsulating phytoconstituents within various nanocarriers including polymeric nanoparticles, lipidic nanoparticles, nanoemulsions, nanogels, gold nanoparticles, and silver nanoparticles. This manuscript emphasizes the recent advancements in phytochemical-loaded nanocarriers efficiently tailored for breast cancer therapy along with patents, current challenges, and future perspectives in this avenue.
Collapse
Affiliation(s)
- Gulshan Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Rohil Panwar
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Sanskriti Saini
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Hardeep Singh Tuli
- Department of Bio-Science and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Rakesh Pahwa
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, Haryana, 136119, India.
| |
Collapse
|
4
|
Al Mashud MA, Kumer A, Jahan I, Somrat MMH, Talukder MEK, Rahman MM, Uddin AFMS, Harun-Or-Rashid M, Rahman MM, Harun-Ur-Rashid M, Shazly GA, Ali Younous Y. Chemoinformatics analysis of Mangifera indica leaves extracted phytochemicals as potential EGFR kinase modulators. Front Chem 2025; 13:1524384. [PMID: 40196294 PMCID: PMC11973374 DOI: 10.3389/fchem.2025.1524384] [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: 11/07/2024] [Accepted: 02/07/2025] [Indexed: 04/09/2025] Open
Abstract
Breast cancer, being among the most frequent and fatal cancers in women, is an enormous issue globally. The critical requirement for novel treatment methods is underscored by its high mortality rate and relentless advancement. Even though breast cancer is one of the world's most common causes of death, the therapeutic avenue is still limited. The aim of this work is to investigate the potential inhibitory effects of specific compounds present in leaf extract from Mangifera indica on the growth of drug-resistant breast cancer protease PDB ID 3w32. The chemical compounds present in Mangifera indica leaves were used to analyze using molecular modeling techniques, such as molecular docking, molecular dynamics (MD) simulations, quantum mechanics (QM) calculations, and the Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) method, in order to examine three key chemical constituents: quercetin (08), catechin (09), and elagic acid (10). The ligands undergo extensive testing to figure out how effective they are against the 3w32-overexpressing breast cancer protein. Quantum calculations retaining HOMO-LUMO analysis might identify important characteristics of molecules, such as chemical potential, electronegativity, hardness, softness, and orbital energy gaps. According to the molecular docking inquiry, ligands 08, 09, and 10 are strong candidates with strong binding affinity for the breast cancer protein that overexpresses 3w32. The protein binding site stability of the chosen natural ligands was verified by MD simulation. These three ligands not only surpass the efficacy of the FDA-approved treatment, but also fulfill the requirements for a possible new inhibitor of breast cancer.
Collapse
Affiliation(s)
- Md. Abdullah Al Mashud
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia, Bangladesh
- Computational Bio-Info Lab, Research and Development Center for Sustainability, Scientific Foundation for Cancer Research, Kushtia, Bangladesh
| | - Ajoy Kumer
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
- Department of Chemistry, College of Arts and Sciences, IUBAT-International University of Business Agriculture and Technology, Dhaka, Bangladesh
| | - Ismat Jahan
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia, Bangladesh
| | - Md. Mehedi Hasan Somrat
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia, Bangladesh
- Computational Bio-Info Lab, Research and Development Center for Sustainability, Scientific Foundation for Cancer Research, Kushtia, Bangladesh
| | - Md. Enamul Kabir Talukder
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Mashiar Rahman
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - A. F. M. Shahab Uddin
- Department of Computer Science and Engineering, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Harun-Or-Rashid
- School of Engineering, Macquarie University Sydney, Sydney, NSW, Australia
- Computational Biology Research Laboratory, Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
| | | | - Mohammad Harun-Ur-Rashid
- Department of Chemistry, College of Arts and Sciences, IUBAT-International University of Business Agriculture and Technology, Dhaka, Bangladesh
| | - Gamal A. Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
5
|
Lambertini M, Jackisch C, Trédan O, Vidal M, Fontes-Sousa M, Valachis A, D'Antona R, Ruz M, Krone E, Brice M, Berjonneau E, Matos S, Dialla O, Guéroult-Accolas L. Patient perception on risk of recurrence and decision-making in the management of HER2-positive early breast cancer: Insights from the ASKHER2 European survey. Breast 2025; 81:104456. [PMID: 40158495 PMCID: PMC11992529 DOI: 10.1016/j.breast.2025.104456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Perceived risk and fear of recurrence in patients with breast cancer (BC) is a matter of concern and may affect their health behaviours and their ability to participate in decision making during their treatment. This survey aimed to examine perceptions and concerns of patients with HER2+ BC. MATERIALS AND METHODS A multi-country, non-interventional, direct-to-patient online survey was conducted between July 22, 2022 and March 1, 2023 in six European countries using a multi-modal recruitment approach. RESULTS Out of 622 included patients, 96.8 % desired involvement in treatment decisions, and 58.5 % felt they had significant influence in the decision-making process. A total of 20.9 % of patients were unaware of their personal risk of recurrence, and 19.5 % reported not discussing this risk with their healthcare providers. The fear of disease recurrence, death, and treatment failure were identified as the most important concerns. Moreover, 30.4 % perceived they had clear communication with healthcare providers on risk of recurrence. A total of 64.5 % were willing to take extra treatments, 60.2 % to undergo more surgery to reduce recurrence risk and 68.5 % were willing to accept further treatments even if recurrence risk decreased by less than 50 %. CONCLUSION Results of this multinational direct-to-patient study examining the perceptions and concerns of women with HER2+ breast cancer underscore the need for physicians to proactively involve patients in their decision-making process, enabling them to participate in a patient-centred approach during treatment decisions.
Collapse
Affiliation(s)
- Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, 16132, Italy.
| | | | - Olivier Trédan
- Centre Léon Bérard, France; Cancer Research Center of Lyon (UMR Inserm 1052 - CNRS 5286), France.
| | | | - Mário Fontes-Sousa
- CUF Tejo, Lisboa, Portugal; Hospital S. Francisco Xavier, ULSLO, Lisboa, Portugal.
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Hsu CY, Rab SO, Zwamel AH, Oghenemaro EF, Chandra M, Rajotiya S, Hjazi A, Prasad K, Atteri S, Chauhan AS. From diagnosis to therapy: The role of LncRNA GAS5 in combatting some cancers affecting women. Gene 2025; 941:149217. [PMID: 39756550 DOI: 10.1016/j.gene.2025.149217] [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: 08/28/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Long non-coding RNAs (lncRNAs) are a collection of non-coding RNA molecules that consist of more than 200 nucleotides. In human malignancies, these lncRNAs exhibit abnormal expression patterns and play a significant role in either suppressing or promoting tumor growth. They achieve this by modulating various functions and mechanisms within cancer cells, including proliferation, invasion, metastasis, apoptosis, and resistance to different therapeutic approaches. The downregulation of long non-coding RNA growth arrest‑specific transcript 5 (GAS5) has been observed in multiple tumor types, indicating its role as a tumor suppressor in cancer. GAS5 exhibits interactions with various proteins, DNA, and microRNAs (miRNAs), leading to the upregulation of several mRNAs encoding suppressor proteins like PTEN. Consequently, this upregulation inhibits tumor growth. In this review, we have examined the existing literature concerning the expression of GAS5 and its diagnostic significance in female tissue-specific cancers, including breast, cervical, ovarian, and endometrial cancers. Additionally, we have explored its interactions with different miRNAs and its impact on cancer progression and resistance to therapy in these malignancies.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ 85004, USA
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq.
| | - Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, PMB 1, Abraka, Delta State, Nigeria
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics,Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat 360003, India
| | - Sumit Rajotiya
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Princse Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Kdv Prasad
- Symbiosis Institute of Business Management, Hyderabad; Symbiosis International (Deemed University), Pune, India
| | - Shikha Atteri
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, Punjab 140307, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| |
Collapse
|
7
|
García-Molina J, Saiz-Vázquez O, Santamaría-Vázquez M, Ortiz-Huerta JH. Efficacy of a Supervised Exercise Program on Pain, Physical Function, and Quality of Life in Patients With Breast Cancer: Protocol for a Randomized Clinical Trial. JMIR Res Protoc 2025; 14:e63891. [PMID: 40073395 PMCID: PMC11947629 DOI: 10.2196/63891] [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: 07/05/2024] [Revised: 09/30/2024] [Accepted: 01/10/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Breast cancer is the second most common cancer in women worldwide. Treatments for this disease often result in side effects such as pain, fatigue, loss of muscle mass, and reduced quality of life. Physical exercise has been shown to effectively mitigate these side effects and improve the quality of life in patients with breast cancer. OBJECTIVE This randomized clinical trial aims to evaluate the efficacy of a 12-week supervised exercise program on pain, physical function, and quality of life in female patients with cancer. METHODS This randomized, double-blind clinical trial will recruit 325 participants, divided into an intervention group receiving the exercise program and a control group receiving standard care recommendations. Outcome measures, including pain (assessed via the Brief Pain Inventory), physical function (Disability of the Arm, Shoulder, and Hand Questionnaire), and quality of life (European Organization for Research and Treatment of Cancer QLQ-C30 and European Organization for Research and Treatment of Cancer QLQ-BR23), will be evaluated at baseline, immediately post intervention, and 12 weeks post intervention. Statistical analysis will involve repeated measures of ANOVA and MANOVA to determine the significance of the intervention's effects across time points. RESULTS Recruitment and data collection will commence in February of 2025, and data analysis is scheduled for completion at the end of 2025. No results are currently available. CONCLUSIONS Physical exercise is anticipated to play a significant role in alleviating pain, enhancing physical function, and improving the quality of life in female patients with cancer. This study will provide robust evidence to support the integration of supervised exercise into standard care protocols for this population. TRIAL REGISTRATION ClinicalTrials.gov NCT06618690; https://clinicaltrials.gov/ct2/show/NCT06618690. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/63891.
Collapse
Affiliation(s)
- Jennifer García-Molina
- Paseo de los Encomendadores, Faculty of Health Sciences, University of Burgos, Burgos, Spain
| | - Olalla Saiz-Vázquez
- Paseo de los Encomendadores, Faculty of Health Sciences, University of Burgos, Burgos, Spain
| | | | | |
Collapse
|
8
|
Thomas J, Malla L, Shibwabo B. Advances in analytical approaches for background parenchymal enhancement in predicting breast tumor response to neoadjuvant chemotherapy: A systematic review. PLoS One 2025; 20:e0317240. [PMID: 40053513 PMCID: PMC11888135 DOI: 10.1371/journal.pone.0317240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/25/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Breast cancer (BC) continues to pose a substantial global health concern, necessitating continuous advancements in therapeutic approaches. Neoadjuvant chemotherapy (NAC) has gained prominence as a key therapeutic strategy, and there is growing interest in the predictive utility of Background Parenchymal Enhancement (BPE) in evaluating the response of breast tumors to NAC. However, the analysis of BPE as a predictive biomarker, along with the techniques used to model BPE changes for accurate and timely predictions of treatment response presents several obstacles. This systematic review aims to thoroughly investigate recent advancements in the analytical methodologies for BPE analysis, and to evaluate their reliability and effectiveness in predicting breast tumor response to NAC, ultimately contributing to the development of personalized and effective therapeutic strategies. METHODS A comprehensive and structured literature search was conducted across key electronic databases, including Cochrane Database of Systematic Reviews, Google Scholar, PubMed, and IEEE Xplore covering articles published up to May 10, 2024. The inclusion criteria targeted studies focusing on breast cancer cohorts treated with NAC, involving both pre-treatment and at least one post-treatment breast dynamic contrast-enhanced Magnetic Resonance Imaging (DCE-MRI) scan, and analyzing BPE utility in predicting breast tumor response to NAC. Methodological quality assessment and data extraction were performed to synthesize findings and identify commonalities and differences among various BPE analytical approaches. RESULTS The search yielded a total of 882 records. After meticulous screening, 78 eligible records were identified, with 13 studies ultimately meeting the inclusion criteria for the systematic review. Analysis of the literature revealed a significant evolution in BPE analysis, from early studies focusing on single time-point BPE analysis to more recent studies adopting longitudinal BPE analysis. The review uncovered several gaps that compromise the accuracy and timeliness of existing longitudinal BPE analysis methods, such as missing data across multiple imaging time points, manual segmentation of the whole-breast region of interest, and over reliance on traditional statistical methods like logistic regression for modeling BPE and pathological complete response (pCR). CONCLUSION This review provides a thorough examination of current advancements in analytical approaches for BPE analysis in predicting breast tumor response to NAC. The shift towards longitudinal BPE analysis has highlighted significant gaps, suggesting the need for alternative analytical techniques, particularly in the realm of artificial intelligence (AI). Future longitudinal BPE research work should focus on standardization in longitudinal BPE measurement and analysis, through integration of deep learning-based approaches for automated tumor segmentation, and implementation of advanced AI technique that can better accommodate varied breast tumor responses, non-linear relationships and complex temporal dynamics in BPE datasets, while also handling missing data more effectively. Such integration could lead to more precise and timely predictions of breast tumor responses to NAC, thereby enhancing personalized and effective breast cancer treatment strategies.
Collapse
Affiliation(s)
- Julius Thomas
- School of Computing and Engineering Sciences, Strathmore University, Nairobi, Kenya
| | - Lucas Malla
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Benard Shibwabo
- School of Computing and Engineering Sciences, Strathmore University, Nairobi, Kenya
| |
Collapse
|
9
|
Gamal NK, El-Naga RN, Ayoub IM, George MY. Neuromodulatory effect of troxerutin against doxorubicin and cyclophosphamide-induced cognitive impairment in rats: Potential crosstalk between gut-brain and NLRP3 inflammasome axes. Int Immunopharmacol 2025; 149:114216. [PMID: 39919456 DOI: 10.1016/j.intimp.2025.114216] [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: 05/16/2024] [Revised: 08/29/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
"Chemobrain" refers to the cognitive impairment induced by chemotherapy. The doxorubicin and cyclophosphamide cocktail has been used for various cancers, especially breast cancer. However, both have been linked to chemobrain as well as gastrointestinal toxicity. Despite being distinct organs, the gut and the brain have a bidirectional connection between them known as the gut-brain axis. This research aimed to study the neuroprotective effect of troxerutin, a rutin derivative, in chemobrain induced by doxorubicin and cyclophosphamide via a potential impact on the gut-inflammasome-brain axis. Troxerutin was administered at 75, 150, and 300 mg/kg doses. Furthermore, behavioral, histological, and acetylcholinesterase assessments were performed. Accordingly, the highest dose of troxerutin was selected to investigate the potential underlying mechanisms. Troxerutin treatment reversed the chemotherapy-fecal metabolite alterations. Additionally, troxerutin demonstrated positive effects against deterioration of intestinal integrity, permeability, and microbial endotoxins translocation, as evidenced by its effect on tight junction proteins; ZO-1, and claudin-1 expression, and lipopolysaccharide serum levels. Consequently, troxerutin hindered lipopolysaccharide-induced oxidative damage, systemic inflammation, and neuroinflammation. Moreover, troxerutin demonstrated antioxidant effects via its impact on lipid peroxidation, catalase levels, and the Nrf2/HO-1 pathway. Furthermore, chemotherapy-induced inflammation was opposed by troxerutin via downregulation of NLRP3, caspase-1, and the downstream cytokines; IL-18 and IL-1β. Importantly, troxerutin did not abrogate the anticancer activity of doxorubicin and cyclophosphamide in human MCF7 cells. Collectively, our study suggested the potentiality of troxerutin as a therapeutic choice against chemobrain by inhibiting the gut-inflammasome-brain axis and hindering acetylcholinesterase, oxidative stress, and neuroinflammation.
Collapse
Affiliation(s)
- Nada K Gamal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University 11566 Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University 11566 Cairo, Egypt
| | - Iriny M Ayoub
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo 11566 Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University 11566 Cairo, Egypt.
| |
Collapse
|
10
|
Li X, Yin J, Song Q, Yang Q, Li C, Gao H. The novel ginseng Rh2 derivative 2-deoxy-Rh2, exhibits potent anticancer effect via the AMPK/mTOR/autophagy signaling pathway against breast cancer. Chem Biol Interact 2025; 409:111422. [PMID: 39961461 DOI: 10.1016/j.cbi.2025.111422] [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: 07/28/2024] [Revised: 11/15/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Breast cancer is the most prevalent cancer and the second leading cause of cancer-related mortality among women globally, resulting in considerable psychological and physical distress for patients. Our previous study synthesized a novel derivative, 2-Deoxy-Rh2, which exhibited anticancer properties by influencing glycolysis and mitochondrial respiration. The objective of the current study was to investigate the anti-proliferative effects and underlying mechanisms of 2-Deoxy-Rh2 on human breast cancer cell lines MCF-7 and MDA-MB-231. In our experiments, we observed that 2-Deoxy-Rh2 reduced cell viability and induced cell cycle arrest, reactive oxygen species accumulation, and mitochondrial dysfunction. Furthermore, treatment with 2-Deoxy-Rh2 affected autophagic flux and induction, leading to increased expression of microtubule-associated protein light chain 3B (LC3B) and decreased expression of sequestosome 1 (P62) expression in both two breast cancer cell lines, which could be reversed by 3-Methyladenine (3-MA). Additionally, the AMPK signaling pathway plays a crucial role in 2-Deoxy-Rh2-induced autophagy. 2-Deoxy-Rh2 modulated the expression levels of mTOR and AMPK in MCF-7 and MDA-MB-231 cells, resulting in the cellular homeostasis disruption, autophagy and apoptosis, which was further corroborated by compound C (CC). Finally, the study validated the antitumor activity and mechanism of 2-Deoxy-Rh2 in vivo using Balb/c mice bearing 4T1 tumor cells. Overall, the results suggest that 2-Deoxy-Rh2 can induce apoptosis and autophagic cell death through the AMPK/mTOR signaling pathway, positioning it as a promising candidate for an antitumor agent against breast cancer.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Radiology, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jianyuan Yin
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, China
| | - Qing Song
- Department of Radiology, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Qi Yang
- Department of Radiology, the First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Chenchen Li
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, China; State Key Laboratory of Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510180, China.
| | - Huan Gao
- Department of Clinical Pharmacy, the First Hospital of Jilin University, Changchun, Jilin, 130021, China; Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
11
|
Hashemkhani M, Sennaroğlu A, Acar HY. ICG-ALA complex for improved phototherapy of cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 264:113121. [PMID: 39923641 DOI: 10.1016/j.jphotobiol.2025.113121] [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: 09/23/2024] [Revised: 01/07/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
5-Aminolevulinic acid (ALA) based photodynamic therapy (PDT) is a clinically approved therapeutic method for cancer treatment. Indocyanine green (ICG) is on the other hand an FDA-approved fluorescent dye that has been widely used in medical imaging in the near-infrared (NIR), and lately recognized as an agent to induce photothermal therapy (PTT). However, the hydrophilicity of ALA and rapid degradation of ICG in aqueous or physiological media as well as their instability limit their clinical application. Besides, the combination of PDT and PTT is a promising alternative to a single therapy approach. Herein, electrostatic binding of ALA to ICG is proposed to bypass such handicaps and provide enhanced therapeutic outcomes with simultaneous PDT and PTT combination. ICG-ALA exhibited excellent biocompatibility up to 50 μg ICG/mL-10 mM ALA in the dark in both SKBR3 and MDA-MB-231 cell lines with higher cell uptake compared to free ALA or ICG. ICG-ALA treatment coupled with 640/808 nm 5 min co-irradiation caused significantly stronger phototoxicity in both cancer cell lines at very low concentrations, reaching near complete loss of viability at 2.5 μg ICG /mL-0.5 mM ALA equivalent concentration of the ICG-ALA. The temperature increase observed during irradiation of the cells and the elevated oxidative stress resulting in the release of caspase 3/7 agrees well with the onset of PTT and PDT. In addition, ICG-ALA demonstrates visualization of cancer cells in both NIR (ICG) and visible (PpIX) regions allowing imaging-guided phototherapy.
Collapse
Affiliation(s)
- Mahshid Hashemkhani
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Université Paris Cité, Matiére et Systémes Complexes MSC, 45 rue des Saints Péres, 75006 Paris, France.
| | - Alphan Sennaroğlu
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Departments of Physics and Electrical-Electronics Engineering, Koç University, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Koç University, Surface Science and Technology Center (KUYTAM), Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey
| | - Havva Yağci Acar
- Koç University, Graduate School of Materials Science and Engineering, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey; Koç University, Department of Chemistry, Rumelifeneri Yolu, Sariyer, 34450 Istanbul, Turkey.
| |
Collapse
|
12
|
Kaya Çakir H, Eroğlu O. Investigation of the synergic effect of mocetinostat and capecitabine in a triple-negative breast neoplasms mouse model. J Investig Med 2025; 73:320-327. [PMID: 39690705 DOI: 10.1177/10815589241309603] [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: 12/19/2024]
Abstract
Combined administration of two or more drugs is emerging as a new strategy in triple-negative breast neoplasms. This is the first study to investigate the combination of the histone deacetylase inhibitor mocetinostat and the antimetabolite drug capecitabine in triple-negative mammary neoplasms in a preclinical mouse model. Thirty-five female mice were grouped into the control group, capecitabine group, mocetinostat group, and combined drugs group. At the end of the experimental period, body weight, and tumor weight were measured and tumor tissue and lung tissue were histologically examined. The results showed that the body weight of mice in the drug-treated groups was reduced by about 18%. Tumor weights were also reduced by 21% in the mocetinostat group, 27.5% in the capecitabine group, and 45% in the combined group. The combination of mocetinostat and capecitabine decreased the formation of tumors and metastases in lung tissue. In summary, the combination of mocetinostat and capecitabine was more effective than either drug alone in reducing the size of triple-negative breast neoplasms in a mouse model.
Collapse
Affiliation(s)
- Hacer Kaya Çakir
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Onur Eroğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Seyh Edebali University, Bilecik, Turkey
| |
Collapse
|
13
|
Fang W, He J, Wang Y, Zhu Y, Qian X, Su D, Gong J, Shang J, He Y, Wu H, Li X. Impact of the national health insurance coverage policy on mecapegfilgrastim utilization for chemoradiotherapy-induced neutropenia in cancer patients in China: a retrospective real-world analysis. Front Pharmacol 2025; 16:1546261. [PMID: 40034821 PMCID: PMC11872703 DOI: 10.3389/fphar.2025.1546261] [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: 12/16/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Objective This study aimed to evaluate mecapegfilgrastim utilization for the prophylaxis of chemotherapy-induced neutropenia in cancer patients and to assess changes caused by the National Health Insurance Coverage (NHIC) policy. Methods Individual patient data, including demographics, medical insurance status, cancer type, and tumor stage, were extracted from electronic medical records in an oncology specialty tertiary hospital in Jiangsu Province, China. An interrupted time series (ITS) analysis with a segmented regression model was applied to evaluate the NHIC policy's effects, and multivariate binary logistic regression analysis was used to identify key factors influencing mecapegfilgrastim utilization. Results The proportion of cancer patients receiving mecapegfilgrastim increased from 8.17% before the NHIC policy implementation to 36.05% after its implementation (P < 0.001). Utilization rose abruptly following the policy intervention (β = 0.143, P < 0.001) and continued to increase significantly afterward (β = 0.011, P = 0.004). However, inequities were observed in mecapegfilgrastim usage among patient subgroups, with utilization closely associated with patients' location, cancer type, and tumor stage after the policy implementation. Conclusion The NHIC policy significantly increased mecapegfilgrastim utilization, enabling more cancer patients to access this medication and effectively benefiting them. To address persistent inequities, the government should consider introducing additional measures, such as increasing the insurance reimbursement cap and separating the cost of expensive innovative anticancer medicines from hospital medical insurance budgets.
Collapse
Affiliation(s)
- Wenqing Fang
- Department of Medical Ethics Supervision, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jizhong He
- Institute of Medical Humanities, Nanjing Medical University, Nanjing, China
- School of Marxism, Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Department of Infection Management, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yulei Zhu
- Office of Scientific Research, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaodan Qian
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Dan Su
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jinhong Gong
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Jingjing Shang
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yuan He
- Institute of Medical Humanities, Nanjing Medical University, Nanjing, China
- School of Marxism, Nanjing Medical University, Nanjing, China
- Laboratory for Digital Intelligence and Health Governance, Nanjing Medical University, Nanjing, China
| | - Hong Wu
- Department of Pharmacy, The Second People’s Hospital of Lianyungang, Lianyungang, China
| | - Xin Li
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Department of Health Policy, School of Health Policy and Management, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Liu X, Wang Y, Wu H, Wang D, Yao H, Ren Z, Cao Y, Cong H, Yu B. Natural polysaccharide hydrogel delivery system remodeling tumor microenvironment to promote postoperative tumor therapy. Int J Biol Macromol 2025; 291:139137. [PMID: 39725109 DOI: 10.1016/j.ijbiomac.2024.139137] [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: 10/23/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
In recent years, postoperative tumor therapy with a suitable approach has been an important issue. Remodeling the tumor microenvironment and accelerating tissue repair can accelerate patients' surgical site recovery, reduce patient pain as well as prevent postoperative tumor recurrence. The shape non-adaptability, cytotoxicity, and non-degradability of some hydrogels still hinder the application of hydrogel-based drug delivery systems in postoperative recovery. Natural polysaccharides (e.g., chitosan, sodium alginate, and hyaluronic acid) are multifunctional compounds with biomimetic advantages to meet the growing demand for nontoxic, targeted therapeutic, and restorative preventive therapies. In this paper, we comprehensively and systematically investigated the synthesis methods, properties, and applications of natural polysaccharide hydrogel (NPH) delivery systems, as well as the mechanisms of remodeling the tumor microenvironment. We aim to provide insights into the design of NPH delivery systems. On this basis, future research directions for NPH delivery systems and their role in remodeling the tumor microenvironment and accelerating postoperative tumor therapy are proposed, and strategies for remodeling the tumor microenvironment using hydrogel delivery systems are discussed, as well as the latest research methods.
Collapse
Affiliation(s)
- Xin Liu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yumei Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Han Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Dayang Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Huanchen Yao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Zekai Ren
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yang Cao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
15
|
Entezam M, Bagheri N, Soltani A, Hosseini SA, Khosravian P, Ferns GA, Ghatrehsamani M. Enhanced antitumor immunity in breast cancer: Synergistic effects of ADAM10/ADAM17 inhibition, metabolic modulation, and camptothecin-loaded selenium nanoparticles. Int J Pharm 2025; 669:125037. [PMID: 39675534 DOI: 10.1016/j.ijpharm.2024.125037] [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: 08/15/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND In this study, we investigate the impact of a multi-targeted therapeutic approach that includes camptothecin (CPT), a potent chemotherapeutic topoisomerase inhibitor; metformin (Met), a metabolic modulator with emerging anti-tumor effects; and GW280264X, an inhibitor of ADAM 10/ADAM 17 enzymes, which are associated with tumor invasion and immune response. The study aims to assess the combined effects of these agents in enhancing CD8+ T cell-mediated anti-tumor immunity and suppressing cancer cell growth in triple-negative breast cancer (TNBC) models, both in vitro and in vivo. METHODS Cell viability was performed on the 4 T1 human TNBC cell line. Furthermore, we examined c-MYC protein expression by western blot, TOX and NR4A expression by Real-time PCR, and the number of CD8+ CD28+ T cells by immunofluorescence assay to demonstrate the anticancer effects of combined of CPT, Met and GW280264X in BC growth, exhaustion and senescence of T cells. RESULTS Regarding cell viability, HA-Se@CPT + Met and HA-Se@CPT + Met + GW280264X treatments decreased 4 T1 cell growth (p < 0.001). Combination therapy of Met, HA-Se@CPT, and GW280264X significantly reduced tumor volume and weight in vivo. This treatment also increased the number of CD8+ CD28+ T cells in the tumor microenvironment (TME) of BC (p < 0.0001) and decreased the expression of TOX and NR4A (p < 0.0001, p < 0.01). Furthermore, decreased expression of c-MYC as an oncogene protein was seen in the single and combined treatment by HA-Se@CPT and GW280264X (p < 0.05). CONCLUSION These findings suggest that of HA-Se@CPT, Met, and GW280264X may inhibit tumor progression in BC by improving the function and infiltration of CD8+ T cells. Their effect is more pronounced when used in combination.
Collapse
Affiliation(s)
- Mahshad Entezam
- Department of Microbiology and Immunology, Shahrekord University of Medical Sciences, Shahrekord, Iran; Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sayedeh Azimeh Hosseini
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Pegah Khosravian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Mahdi Ghatrehsamani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
16
|
Dabi A, Brown JS, Gatenby RA, Jones CD, Schrider DR. Evolutionary rescue model informs strategies for driving cancer cell populations to extinction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625315. [PMID: 39651238 PMCID: PMC11623570 DOI: 10.1101/2024.11.26.625315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Cancers exhibit a remarkable ability to develop resistance to a range of treatments, often resulting in relapse following first-line therapies and significantly worse outcomes for subsequent treatments. While our understanding of the mechanisms and dynamics of the emergence of resistance during cancer therapy continues to advance, questions remain about how to minimize the probability that resistance will evolve, thereby improving long-term patient outcomes. Here, we present an evolutionary simulation model of a clonal population of cells that can acquire resistance mutations to one or more treatments. We leverage this model to examine the efficacy of a two-strike "extinction therapy" protocol, in which two treatments are applied sequentially to first contract the population to a vulnerable state and then push it to extinction, and compare it to a combination therapy protocol. We investigate how factors such as the timing of the switch between the two strikes, the rate of emergence of resistant mutations, the doses of the applied drugs, the presence of cross-resistance, and whether resistance is a binary or a quantitative trait affect the outcome. Our results show that the timing of switching to the second strike has a marked effect on the likelihood of driving the cancer to extinction, and that extinction therapy outperforms combination therapy when cross-resistance is present. We conduct an in silico trial that reveals when and why a second strike will succeed or fail. Finally, we demonstrate that our conclusions hold whether we model resistance as a binary trait or as a quantitative, multi-locus trait.
Collapse
Affiliation(s)
- Amjad Dabi
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joel S. Brown
- Department of Cancer Biology and Evolution, Moffitt Cancer Center, Tampa, FL, USA
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Robert A. Gatenby
- Department of Cancer Biology and Evolution, Moffitt Cancer Center, Tampa, FL, USA
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Diagnostic Imaging Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Corbin D. Jones
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, USA
- Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, North Carolina, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Daniel R. Schrider
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
17
|
Englund E, Strandéus M, Engvall K, Oliva D. Neutropenic event incidence in women with early-stage breast cancer receiving neoadjuvant or adjuvant chemotherapy: a retrospective study. BMJ Support Palliat Care 2025:spcare-2024-005329. [PMID: 39793979 DOI: 10.1136/spcare-2024-005329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVES To assess the incidence of neutropenia, febrile neutropenia, documented infection with neutropenia and fever associated with early-stage breast cancer (BC) in a real-life setting. METHODS A retrospective study that includes 88 women with BC who received a first dose of Epirubicin plus Cyclophosphamide with or without 5-Fluorouracil, in the county hospital of Ryhov, Sweden. The patients were included continuously from May 2017 to November 2020 and were ≥18 years old. All data was collected in a form and the G-CSF prophylaxis was checked to ensure that it was given during the treatments. RESULTS The median age among the patients was 59 (min 27-82max) years. 79 patients were treated with primary prophylaxis with Granulocyte-colony stimulating factor (G-CSF) support. Seven (8.0%) patients were affected by a neutropenic episode, including one (1.1%) patient with FN and two (2.3%) patients with documented infection (online supplemental table 1). CONCLUSION The incidence of neutropenic events in this study is relatively low. A higher incidence of neutropenic episodes is observed in patients ≥ 60 years old compared with younger patients, despite lower doses of chemotherapy for the elderly. The use of G-CSF in the elderly, regardless of the dose of chemotherapy, may be needed to decrease the incidence of neutropenia and its consequences. The adverse effects of G-CSF and its cost-effectiveness are important perspectives which should be included in the treatment.
Collapse
Affiliation(s)
- Elin Englund
- Department of Oncology, County Hospital Ryhov, Jönköping, Jonkoping County, Sweden
| | - Michael Strandéus
- Department of Oncology, County Hospital Ryhov, Jönköping, Jonkoping County, Sweden
| | - Kristina Engvall
- Department of Oncology, County Hospital Ryhov, Jönköping, Jonkoping County, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linkoping, Sweden
| | - Delmy Oliva
- Department of Oncology, County Hospital Ryhov, Jönköping, Jonkoping County, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linkoping, Sweden
| |
Collapse
|
18
|
Guedes AR, Soares JP, Cunha R, Silva AM, Gaivão I. DNA damage of peripheral blood lymphocytes as a dual biomarker: Diagnostic and treatment response in woman breast cancer patients. Cancer Biomark 2025; 42:18758592241308748. [PMID: 40109217 DOI: 10.1177/18758592241308748] [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: 03/22/2025]
Abstract
BackgroundBreast cancer is the leading malignancy among women and the lack of ideal early biomarkers hampers diagnosis and treatment monitoring. Genomic instability, central to breast cancer development, makes DNA damage a potential biomarker for these purposes.ObjectiveThis study aims to evaluate the predictive value of DNA damage for diagnosis, and treatment monitoring in breast cancer, with CA 15-3, a conventional cancer biomarker, included for comparison to assess the added value of DNA damage measurement.MethodsDNA damage was measured in peripheral blood lymphocytes of 58 breast cancer patients, and 31 healthy controls, employing comet assay, both before and after treatment. Serum CA 15-3 levels were assessed at the same time points for comparison.ResultsDNA damage levels were significantly higher in cancer patients compared to healthy controls, with the most elevated levels observed in patients with advanced-stage disease, irrespective of age, sex, lifestyle, or genetic status. Post-treatment assessments showed a significant rise in DNA damage. In comparison, CA 15-3 showed less consistent relevance for diagnostic and monitoring.ConclusionsThis study underscores the greater potential of DNA damage as a consistent and reliable biomarker for breast cancer, with CA 15-3 providing complementary but less consistent data for clinical decision-making.
Collapse
Affiliation(s)
- Ana Rita Guedes
- Animal and Veterinary Research Center (CECAV) and Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), UTAD, Vila Real, Portugal
- Department of Biology and Environment (DeBA) and Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), UTAD, Vila Real, Portugal
| | - Jorge Pinto Soares
- Research Centre of Sports, Health, and Human Development (CIDES), UTAD, Vila Real, Portugal
| | - Renato Cunha
- Local Health Unit of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Amélia Maria Silva
- Department of Biology and Environment (DeBA) and Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), UTAD, Vila Real, Portugal
| | - Isabel Gaivão
- Animal and Veterinary Research Center (CECAV) and Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), UTAD, Vila Real, Portugal
| |
Collapse
|
19
|
Jiang J, Shi S, Zhang W, Li C, Sun L, Ge Q, Li X. Circ_RPPH1 facilitates progression of breast cancer via miR-1296-5p/TRIM14 axis. Cancer Biol Ther 2024; 25:2360768. [PMID: 38816350 PMCID: PMC11141472 DOI: 10.1080/15384047.2024.2360768] [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: 12/18/2023] [Accepted: 05/23/2024] [Indexed: 06/01/2024] Open
Abstract
Circular RNA Ribonuclease P RNA Component H1 (circ_RPPH1) and microRNA (miRNA) miR-1296-5p play a crucial role in breast cancer (BC), but the molecular mechanism is vague. Evidence showed that miR-1296-5p can activate tripartite motif-containing 14 (TRIM14). Clinical indications of eighty BC patients were collected and the circ_RPPH1 expression was detected using real-time quantitative PCR. MCF-7 and MDA-MB-231 cells were transfected with overexpression or knockdown of circ_RPPH1, miR-1296-5p, or TRIM14. Cell counting kit-8, cell cloning formation, wound healing, Transwell, and flow cytometry assays were performed to investigate the malignant phenotype of BC. The dual-luciferase reporter gene analyses were applied to reveal the interaction between these target genes. Subcutaneous tumorigenic model mice were established with circ_RPPH1 overexpression MDA-MB-231 cells in vivo; the tumor weight and volume, levels of miR-1296-5 and TRIM14 mRNA were measured. Western blot and immunohistochemistry were used to detect TRIM14 in cells and mice. Circ_RPPH1 levels were notably higher in BC patients and have been found to promote cell proliferation, invasion, and migration of BC cells. Circ_RPPH1 altered cell cycle and hindered apoptosis. Circ_RPPH1 knockdown or miR-1296-5p overexpression inhibited the malignant phenotype of BC. Furthermore, miR-1296-5p knockdown reversed circ_RPPH1's promotion effects on BC. Interestingly, TRIM14 overexpression counteracts the inhibitory effects of miR-1296-5p overexpression and circ_RPPH1 silencing on BC. Moreover, in BC tumor-bearing mice, circ_RPPH1 overexpression led to increased TRIM14 expression and facilitated tumor growth. Circ_RPPH1 enhanced BC progression through miR-1296-5p/TRIM14 axis, indicating its potential as a biomarker and therapeutic target in BC.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Shenghong Shi
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Wei Zhang
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Chao Li
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Long Sun
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Qidong Ge
- Department of Breast Surgery, Ningbo, Zhejiang, China
| | - Xujun Li
- Department of Breast Surgery, Ningbo, Zhejiang, China
| |
Collapse
|
20
|
Parviainen E, Nurmenniemi S, Ravaioli S, Bravaccini S, Manninen A, Jukkola A, Selander K. Human papillomavirus E6 alters Toll-like receptor 9 transcripts and chemotherapy responses in breast cancer cells in vitro. Mol Biol Rep 2024; 52:43. [PMID: 39644451 PMCID: PMC11625066 DOI: 10.1007/s11033-024-10143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Toll-like receptor 9 (TLR9) is a DNA recognizing receptor expressed also in several cancers. Decreased TLR9 expression is associated with poor prognosis in triple negative breast cancer (TNBC), but the role of TLR9 in breast cancer pathophysiology is currently unclear. Regulation of TLR9 expression in breast cancer is poorly understood. Human papillomavirus (HPV) infections suppress TLR9 expression in cervical cancers but the association between HPV and breast cancer has remained controversial. The aim of this study was to test if HPV16 can suppress TLR9 expression in breast cancer cells and affect cell behavior. METHODS AND RESULTS Human T-47D and MDA-MB-231 breast cancer cells were transduced with lentivirus encoding HPV16 E6 oncoprotein. The effects of E6 on TLR9 mRNA and protein expression, and cell proliferation, migration, invasion and sensitivity to chemotherapy were studied in vitro. Breast cancer tissue samples (n = 37) were analyzed for the presence of HPV DNA. E6 expression decreased TLR9 mRNA expression in MDA-MB-231 and T-47D cells in hypoxia. E6 expression altered breast cancer cell proliferation and made cells significantly less sensitive to the growth inhibitory effects of chemotherapeutic agents. HPV L1 gene was not detected in a small pilot cohort of clinical breast cancer specimens. CONCLUSION HPV16 may influence breast cancer cell TLR9 transcription and chemotherapy responses and could thereby affect breast cancer prognosis. These results suggest that HPV may have a previously unrecognized role in breast cancer pathophysiology and warrant further studies on the topic.
Collapse
Affiliation(s)
- Essi Parviainen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Sini Nurmenniemi
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland.
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland.
| | - Sara Ravaioli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
- Faculty of Medicine and Surgery, "Kore" University of Enna, Enna, Italy
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Arja Jukkola
- Department of Oncology and Radiotherapy, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere Cancer Center, Tampere University, Tampere, Finland
| | - Katri Selander
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland
- Department of Oncology and Radiation Therapy, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
21
|
Urtekin D, Eroglu SA. Effect of training based on Orem's self-care deficit theory on breast cancer patients' management of chemotherapy-related side effects and self-care behaviors: A randomized controlled trial. Eur J Oncol Nurs 2024; 73:102698. [PMID: 39395233 DOI: 10.1016/j.ejon.2024.102698] [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: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024]
Abstract
PURPOSE To examine the effects of training based on Orem's self-care deficit theory on breast cancer patient's physical, social, and psychological well-being and self-care behaviors during chemotherapy. METHODS This randomized controlled trial was based on a pretest-posttest experimental design. The study sample consisted of 62 breast cancer patients (n = 31 for each of the intervention and control groups) receiving cyclophosphamide-epirubicin or cyclophosphamide-adriamycin treatment in the medical oncology department of a university hospital in Tekirdağ, Turkey. The breast cancer patients in the intervention group received training based on Orem's self-care deficit theory regarding physical, social, and psychological well-being and self-care behaviors during chemotherapy. The researcher conducted two face-to-face follow-ups (during the second and third chemotherapy cycles, 3 weeks apart) and a posttest assessment. Pretest and posttest symptom scores and self-care survey results were compared for both groups. RESULTS The Nightingale Symptom Assessment Scale (N-SAS) first follow-up, second follow-up, and posttest scores showed that the intervention group experienced fewer negative changes in quality of life than the control group based on the physical, social, and psychological subdimension and total scale scores (p < .05). The most positive changes in the intervention group's self-care behaviors were observed for items about caring for and protecting hair, using protective gloves when doing chores, monitoring weight, and limiting social meetings when blood values were low to protect against possible infection. CONCLUSION Training based on Orem's self-care deficit theory positively influenced breast cancer patients' management of chemotherapy-related side effects and self-care behaviors. Chemotherapy-related side effects should be evaluated frequently. Moreover, patients' needs should be determined, and training should be tailored to their needs.
Collapse
Affiliation(s)
- Dilek Urtekin
- Tekirdag Namik Kemal University Hospital, Therapeutic Apheresis Center, Tekirdag, Turkey
| | - Semiha Akin Eroglu
- University of Health Sciences Hamidiye Faculty of Nursing, Department of Internal Medicine Nursing, Istanbul, Turkey.
| |
Collapse
|
22
|
Nair A, Singh R, Gautam N, Saxena S, Mittal S, Shah S, Talegaonkar S. Multifaceted role of phytoconstituents based nano drug delivery systems in combating TNBC: A paradigm shift from chemical to natural. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9207-9226. [PMID: 38953968 DOI: 10.1007/s00210-024-03234-0] [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: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
Triple negative breast cancer is considered to be a malignancy of grave concern with limited routes of treatment due to the absence of specific breast cancer markers and ambiguity of other potential drug targets. Poor prognosis and inadequate survival rates have prompted further research into the understanding of the molecular pathophysiology and targeting of the disease. To overcome the recurrence and resistance mechanisms of the TNBC cells, various approaches have been devised, and are being continuously evaluated to enhance their efficacy and safety. Chemo-Adjuvant therapy is one such treatment modality being employed to improve the efficiency of standard chemotherapy. Combining chemo-adjuvant therapy with other upcoming approaches of cancer therapeutics such as phytoconstituents and nanotechnology has yielded promising results in the direction of improving the prognosis of TNBC. Numerous nanoformulations have been proven to substantially enhance the specificity and cellular uptake of drugs by cancer cells, thus reducing the possibility of unintended systemic side effects within cancer patients. While phytoconstituents offer a wide variety of beneficial active constituents useful in cancer therapeutics, most favorable outcomes have been observed within the scope of polyphenols, isoquinoline alkaloids and isothiocyanates. With an enhanced understanding of the molecular mechanisms of TNBC and the advent of newer targeting technologies and novel phytochemicals of medicinal importance, a new era of cancer theranostic treatments can be explored. This review hopes to instantiate the current body of research regarding the role of certain phytoconstituents and their potential nanoformulations in targeting specific TNBC pathways for treatment and diagnostic purposes.
Collapse
Affiliation(s)
- Anandita Nair
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Roshni Singh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Namrata Gautam
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Shilpi Saxena
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Saurabh Mittal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, U.P, Noida, 201303, India.
| | - Sadia Shah
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, 226003, India.
| | - Sushama Talegaonkar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India.
| |
Collapse
|
23
|
Mao C, Fang J, Zou S, Huang Y, Chen X, Ding X, Fang Z, Zhang N, Lou Y, Chen Z, Ding W, Ma Z. Discovery of the First-in-Class Dual-Target ROCK/HDAC Inhibitor with Potent Antitumor Efficacy in Vivo That Trigger Antitumor Immunity. J Med Chem 2024; 67:20619-20638. [PMID: 39523548 DOI: 10.1021/acs.jmedchem.4c02183] [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: 11/16/2024]
Abstract
Triple-negative breast cancer (TNBC) represents a highly aggressive and heterogeneous malignancy. Currently, multitarget drug approaches present a promising therapeutic approach for TNBC. Utilizing a combinatorial chemistry strategy to construct a virtual screening database, dual ROCK/HDAC-targeting benzothiophene compounds were identified. Notably, compound 10h effectively inhibits ROCK1/2 and HDAC1/2/3/6/8 while demonstrating potent antiproliferative activity against breast cancer cells. In an orthotopic mouse model of breast cancer, 10h significantly suppressed tumor growth without apparent toxicity. Importantly, 10h induced immunogenic cell death (ICD), promoted dendritic cells (DCs) maturation, and activated T cells, thereby initiating antitumor immunity. In conclusion, compound 10h is a novel dual-target ROCK/HDAC inhibitor that represents a promising treatment strategy for TNBC.
Collapse
Affiliation(s)
- Churu Mao
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Jiebin Fang
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Shijie Zou
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yun Huang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xiaoming Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xia Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Zhangyun Fang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Ningjing Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yijie Lou
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wanjing Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Zhongjun Ma
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| |
Collapse
|
24
|
Chen R, Zhang H, Zhao X, Zhu L, Zhang X, Ma Y, Xia L. Progress on the mechanism of action of emodin against breast cancer cells. Heliyon 2024; 10:e38628. [PMID: 39524792 PMCID: PMC11550755 DOI: 10.1016/j.heliyon.2024.e38628] [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: 03/08/2024] [Revised: 08/22/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
At present, the role of active ingredients of traditional Chinese medicine in tumor therapy has gradually attracted people's attention, and anthraquinones, which are structurally similar to adriamycin and epirubicin, are one of the hotspots of research. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is a natural anthraquinone compound isolated from rhubarb, Polygonum cuspidatum, and aloe vera. In recent years, emodin has received widespread attention for its remarkable anti-tumor effects, and its anti-breast cancer effects are manifested as induction of apoptosis, inhibition of tumor cell proliferation, inhibition of invasion and metastasis of tumor cells, and anti-tumor drug resistance. Moreover, emodin can act against multiple types of breast cancer cells by acting on different targets. In this paper, we reviewed the latest research progress on the anti-breast cancer effects of emodin and its anti-tumor mechanism, to provide reference and information for the treatment of breast cancer and the development of anti-tumor drugs.
Collapse
Affiliation(s)
- Ruoqing Chen
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, PR China
| | - Xue Zhao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Lin Zhu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - XiaoYu Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Yuning Ma
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| |
Collapse
|
25
|
Guan C, Han Y, Ling Z, Meng X, Zhang B, Dong W, Zhang D, Chen K. Nanomaterials: breaking the bottleneck of breast cancer drug resistance. Front Immunol 2024; 15:1492546. [PMID: 39606228 PMCID: PMC11599193 DOI: 10.3389/fimmu.2024.1492546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Drug resistance poses a significant challenge in the treatment of breast cancer. In recent years, a variety of nanomaterials have been discovered and synthesized that can selectively target tumor cells and play a crucial role in the advancement of breast cancer therapies. As our understanding of tumor heterogeneity deepens, the emerging potential of nanomaterials in addressing drug resistance has garnered considerable attention. These materials not only selectively target tumor cells but also possess unique properties that make them promising options for cancer treatment, including low toxicity, excellent biocompatibility, ease of preparation, the ability to carry antitumor drugs, and customizable surface functions. In this review, we will comprehensively summarize two key developments in breast cancer treatment: the application of antitumor drugs and nanomaterials. We will explore the mechanisms by which nanomaterials improve drug resistance in breast cancer, targeted nanotherapy strategies to mitigate this resistance, and recent research advancements in anticancer nanomaterials. This overview aims to highlight the significant role of nanomaterials in breast cancer treatment and provide a theoretical framework for identifying optimal treatment strategies in the future.
Collapse
Affiliation(s)
- Chao Guan
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Yahao Han
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Zhenzheng Ling
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Xiang Meng
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Baolin Zhang
- The Fourth Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Wanwei Dong
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Di Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Keyan Chen
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Shen R, Zhou J, Xin L, Zhou HB, Huang J. OBHSA, a novel selective estrogen receptor degrader, overcomes tamoxifen resistance through cell cycle arrest and unfolded protein response-mediated apoptosis in breast cancer. J Steroid Biochem Mol Biol 2024; 244:106599. [PMID: 39147211 DOI: 10.1016/j.jsbmb.2024.106599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/23/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Breast cancer (BC) is a highly heterogeneous tumor that has surpassed lung cancer as the most frequently diagnosed cancer in women. In clinical practice,the primary approach for treating estrogen receptor alpha (ERα)-positive BC is through endocrine therapy, which involves targeting the ERα using medications like tamoxifen and fulvestrant. However, the problem of de novo or acquired resistance poses a significant clinical challenge, emphasizing the critical need for the development of novel therapeutic strategies. In this regard, we have successfully designed and developed a novel selective estrogen receptor degrader (SERD) called OBHSA, which specifically targets and degrades ERα, demonstrating remarkable efficacy. Our findings revealed the effectiveness of OBHSA in inhibiting the proliferation of various BC cells, including both tamoxifen-sensitive and tamoxifen-resistant BC cells, indicating its great potential to overcome endocrine resistance. In terms of mechanism, we discovered that OBHSA overcame tamoxifen resistance through two distinct pathways. Firstly, OBHSA degraded cyclin D1 in an ERα-dependent manner, thereby blocking the cell cycle. Secondly, OBHSA induced an elevation in intracellular reactive oxygen species, triggering an excessive activation of the unfolded protein response (UPR) and ultimately leading to apoptotic cell death. In summary, our finding demonstrated that OBHSA exerts anti-tumor effects by inducing cell cycle arrest and UPR-mediated apoptosis. These findings hold promise for the development of novel therapeutic drugs targeting endocrine-resistant BC.
Collapse
Affiliation(s)
- Rong Shen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Bayi Road, Wuhan, Hubei 430072, China
| | - Jiawei Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Bayi Road, Wuhan, Hubei 430072, China
| | - Lilan Xin
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hai-Bing Zhou
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Bayi Road, Wuhan, Hubei 430072, China.
| |
Collapse
|
27
|
Seo JW, Lee YS, Jeon IS, Yu JE, Yoo JS, Koo JK, Son DJ, Yoon JS, Han SB, Yoon DY, Roh YS, Hong JT, Shim JH. IL-32γ Induced Autophagy Through Suppression of MET and mTOR Pathways in Liver Tumor Growth Inhibition. Int J Mol Sci 2024; 25:11678. [PMID: 39519229 PMCID: PMC11547131 DOI: 10.3390/ijms252111678] [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: 09/29/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Interleukin-32γ (IL-32γ) has diverse functions in various malignancies. In this study, we investigated the role of IL-32γ in autophagy induction in liver cancer cells and delineated the underlying mechanisms. We found that the increased IL-32γ expression inhibited the growth, cell cycle progression, and migration of HepG2 and Hep3B cell lines; it also decreased the expression of related proteins. Furthermore, the IL-32γ overexpression induced autophagy, as indicated by the number of puncta, the expression of LC3, and the expression of autophagy-related markers. The expression levels of LAMP1, a protein essential for autophagosome formation, and colocalization with LC3 also increased. Big data analysis revealed that the expression of MET, a well-known target of autophagy, and the expression of mTOR and mTOR-related proteins were decreased by the IL-32γ overexpression. The combination treatment of MET inhibitor, cabozantinib (2 µM), and IL-32γ overexpression further increased the number of puncta, the colocalization of LC3 and LAMP1, and the expression of autophagy-related proteins. In vivo, liver tumor growth was suppressed in the IL-32γ-overexpressing mouse model, and autophagy induction was confirmed by the increased expression of LC3 and LAMP1 and the decreased expression of autophagy pathway markers (MET and mTOR). Autophagy was also decreased in the liver tumor sample of human patients. ROC curve and spearman analysis revealed that the expression levels of LC3 and IL-32γ were significantly correlated in human tumor serum and tissues. Therefore, IL-32γ overexpression induced autophagy in liver tumors through the suppression of MET and mTOR pathways critical for tumor growth inhibition.
Collapse
Affiliation(s)
- Ji-Won Seo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Yong-Sun Lee
- Ministry of Food and Drug Safety, 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju 28159, Chungbuk, Republic of Korea;
| | - In-Sook Jeon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Ji-Eun Yu
- College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea
| | - Jun-Sang Yoo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Ja-Keun Koo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Dong-Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jae-Suk Yoon
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea;
| | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jin-Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si 28160, Chungbuk, Republic of Korea; (J.-W.S.); (I.-S.J.); (J.-S.Y.); (J.-K.K.); (D.-J.S.); (J.-S.Y.); (S.-B.H.); (Y.-S.R.)
| | - Jung-Hyun Shim
- College of Pharmacy, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea;
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Jeonnam, Republic of Korea
| |
Collapse
|
28
|
Orobator ON, Mertens RT, Obisesan OA, Awuah SG. Energy and endoplasmic reticulum stress induction by gold(III) dithiocarbamate and 2-deoxyglucose synergistically trigger cell death in breast cancer. J Biol Chem 2024:107949. [PMID: 39481597 DOI: 10.1016/j.jbc.2024.107949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
The elusiveness of triple-negative breast cancer from targeted therapy has redirected focus towards exploiting the metabolic shortcomings of these highly metastatic subtypes of breast cancer. Cueing from the metabolic heterogeneity of TNBC and the exposition of the dual dependence of some TNBCs on OXPHOS and glycolysis for ATP, we herein report the efficacy of cotreatment of TNBCs with an OXPHOS inhibitor, 2a and 2DG, a potent glycolysis inhibitor. 2a-2DG cotreatment inhibited TNBC cell proliferation with IC50 of ∼5 to 36 times lower than that of 2a alone and over 5000 times lower than IC50 of 2DG alone. 2a-2DG cotreatment suppressed mitochondrial ATP production and significantly induced AMPK activation. Mechanistic studies revealed the distinct yet synergistic contributions of 2a and 2DG to the antiproliferative effect of the cotreatment. While 2a induced apoptotic cell death, 2DG sensitized TNBCs to the antiproliferative effects of 2a via endoplasmic reticulum stress induction. Strikingly, the combination of 2a-2DG ablated SUM159 tumors in an orthotopic xenograft mouse model. This study highlights the synergistic effect of a gold-based complex with 2DG and the potential benefit of multi-metabolic pathways targeting as an effective therapeutic strategy against TNBCs.
Collapse
Affiliation(s)
- Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Oluwatosin A Obisesan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States; Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA; Center for Bioelectronics and Nanomedicine, University of Kentucky, Lexington KY, 40506, USA.
| |
Collapse
|
29
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [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: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
30
|
Esfandyari-Manesh M, Morshedi B, Joolaie P, Dinarvand R. Self-assembled nanoparticles of alginate and paclitaxel-triphenylphosphonium for mitochondrial apoptosis targeting. Med Oncol 2024; 41:299. [PMID: 39443414 DOI: 10.1007/s12032-024-02540-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Paclitaxel (PTX), an antimitotic drug from the taxanes group, prevents the proliferation of breast cancer cells through mitosis arrest and activation by a cascade of signaling pathways that lead to apoptosis. Mitochondria is one of the important signaling routes for inducing apoptosis. For mitochondria targeting, triphenylphosphonium (TPP) with a delocalized charge and hydrophobic nature was utilized as a moiety to facilitate penetration through a phospholipid membrane of mitochondria. PTX-TPP was synthesized via pH-sensitive ester bond between hydroxyl groups of PTX and carboxylic acid of (4-carboxybutyl) TPP. Then PTX-TPP prodrug encapsulated in alginate nanoparticles, which were self-assembled by the ionotropic complexation technique for enhancement of mitochondrial apoptosis in breast cancer cells. The loading of PTX-TPP conjugation in self-assembled alginate nanoparticles was 16.5% and the particle size of nanoparticles was 123 nm with zeta potential around - 25.8 Mv. The in vitro cytotoxicity and IC50 of PTX-TPP nanoparticles in the growth of MCF7 cancer cell increased 6.3-fold higher than free PTX. The early apoptotic cells and the late apoptotic/necrotic cells for PTX-TPP nanoparticles were 11.6 and 3.9-fold higher than free PTX. This study indicated this mitochondrial-targeted self-assembled nanoparticles can inhibit the tumor cell growth of breast cancer.
Collapse
Affiliation(s)
- Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bahar Morshedi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Joolaie
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Debnath A, Mazumder R, Singh RK, Singh AK. Discovery of novel CDK4/6 inhibitors from fungal secondary metabolites. Int J Biol Macromol 2024; 282:136807. [PMID: 39447792 DOI: 10.1016/j.ijbiomac.2024.136807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/03/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The development of targeted therapies for breast cancer, particularly those focusing on cyclin-dependent kinases 4/6 (CDK4/6), has significantly improved patient outcomes. However, the currently approved CDK4/6 inhibitors are associated with various side effects, underscoring the need for novel compounds with enhanced efficacy and safety profiles. This study aimed to identify potential CDK4/6 inhibitors from MeFSAT, a database of fungal secondary metabolites using an in-silico screening approach. The virtual screening process incorporated drug-likeness filters, ADME and toxicity predictions, consensus molecular docking, and 200 ns molecular dynamics simulations. Out of 411 initial compounds, two molecules demonstrated favorable binding interactions and stability with the CDK4/6 protein complex. The MTT assay showed that MSID000025 had dose-dependent cytotoxicity against MCF7 breast cancer cells. This suggests that MSID000025 could be a good candidate CDK4/6 inhibitor for treating breast cancer. Our study highlights the potential of fungal secondary metabolites as a source of novel compounds for drug discovery. It provides a framework for identifying CDK4/6 inhibitors with improved therapeutic properties.
Collapse
Affiliation(s)
- Abhijit Debnath
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida 201306, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida 201306, Uttar Pradesh, India.
| | - Rajesh Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Anil Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
32
|
Santos P, Rezende CP, Piraine R, Oliveira B, Ferreira FB, Carvalho VS, Calado RT, Pellegrini M, Almeida F. Extracellular vesicles from human breast cancer-resistant cells promote acquired drug resistance and pro-inflammatory macrophage response. Front Immunol 2024; 15:1468229. [PMID: 39474419 PMCID: PMC11518763 DOI: 10.3389/fimmu.2024.1468229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/30/2024] [Indexed: 05/01/2025] Open
Abstract
Introduction Breast cancer is a significant public health problem around the world, ranking first in deaths due to cancer in females. The therapy to fight breast cancer involves different methods, including conventional chemotherapy. However, the acquired resistance that tumors develop during the treatment is still a central cause of cancer-associated deaths. One mechanism that induces drug resistance is cell communication via extracellular vesicles (EVs), which can carry efflux transporters and miRNA that increase sensitive cells' survivability to chemotherapy. Methods Our study investigates the transcription changes modulated by EVs from tamoxifen- and doxorubicin-resistant breast cancer cells in sensitive cells and how these changes may induce acquired drug resistance, inhibit apoptosis, and increase survivability in the sensitive cells. Additionally, we exposed human macrophages to resistant EVs to understand the influence of EVs on immune responses. Results Our results suggest that the acquired drug resistance is associated with the ability of resistant EVs to upregulate several transporter classes, which are directly related to the increase of cell viability and survival of sensitive cells exposed to EVs before a low-dose drug treatment. In addition, we show evidence that resistant EVs may downregulate immune system factors to evade detection and block cell death by apoptosis in sensitive breast cancer cells. Our data also reveals that human macrophages in contact with resistant EVs trigger a pro-inflammatory cytokine secretion profile, an effect that may be helpful for future immunotherapy studies. Discussion These findings are the first transcriptome-wide analysis of cells exposed to resistant EVs, supporting that resistant EVs are associated with the acquired drug resistance process during chemotherapy by modulating different aspects of sensitive cancer cells that coffer the chemoresistance.
Collapse
Affiliation(s)
- Patrick Santos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Caroline P. Rezende
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Renan Piraine
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bianca Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francielle B. Ferreira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Vinicius S. Carvalho
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, United States
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
33
|
Wang J, Wang X, Pang Y. StructNet-DDI: Molecular Structure Characterization-Based ResNet for Prediction of Drug-Drug Interactions. Molecules 2024; 29:4829. [PMID: 39459198 PMCID: PMC11510539 DOI: 10.3390/molecules29204829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
This study introduces a deep learning framework based on SMILES representations of chemical structures to predict drug-drug interactions (DDIs). The model extracts Morgan fingerprints and key molecular descriptors, transforming them into raw graphical features for input into a modified ResNet18 architecture. The deep residual network, enhanced with regularization techniques, efficiently addresses training issues such as gradient vanishing and exploding, resulting in superior predictive performance. Experimental results show that StructNet-DDI achieved an AUC of 99.7%, an accuracy of 94.4%, and an AUPR of 99.9%, demonstrating the model's effectiveness and reliability. These findings highlight that StructNet-DDI can effectively extract crucial features from molecular structures, offering a simple yet robust tool for DDI prediction.
Collapse
Affiliation(s)
- Jihong Wang
- School of Computer, Guangdong University of Education, Guangzhou 510310, China
| | - Xiaodan Wang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Yuyao Pang
- School of Pharmaceutical Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| |
Collapse
|
34
|
Li S, FeiyuTeng, Zhang J, Zhang P, Li M, Wang X, Li K. Tanshinone IIA potentiates the chemotherapeutic effect of doxorubicin against breast cancer cells and attenuates the cardiotoxicity of doxorubicin by regulating ERK1/2 pathway. J Biochem Mol Toxicol 2024; 38:e23851. [PMID: 39267350 DOI: 10.1002/jbt.23851] [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: 05/27/2024] [Revised: 08/07/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
Doxorubicin (Dox) is frequently employed as a chemotherapy agent for breast cancer. As the chemotherapy moves forward, breast cancer cells tend to develop resistance to Dox, besides that, Dox are also easy to cause cardiotoxicity related to cumulative dose. Therefore, how to potentiate the chemosensitivity of breast cancer cells to Dox while attenuating its cardiotoxicity has become a research hotspot. Tanshinone IIA (Tan IIA) is known for its anticancer activity as well as for its cardioprotective effects. In view of the aforementioned facts, we assessed whether Tan IIA possesses synergism and attenuation effect on Dox for breast cancer chemotherapy. Our studies in vitro indicated that, Tan IIA could potentiate the effect of Dox on breast cancer cells proliferation inhibition and apoptosis promotion by inhibiting ERK1/2 pathway, but interestingly, Tan IIA attenuated the cytotoxicity of Dox to myocardial cells by activating ERK1/2 pathway. Additionally, our studies in vivo also suggested that Tan IIA potentiated the chemotherapeutic effect of Dox against breast cancer while attenuating Dox-induced myocardial injury. Given that Tan IIA had a synergism and attenuation effect on Dox, we believed that Tan IIA can be used as an ideal drug in combination with Dox for breast cancer therapy.
Collapse
Affiliation(s)
- Shizheng Li
- Department of Emergency Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - FeiyuTeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jianing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Puwei Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Manyuan Li
- Department of Clinical Laboratory, Jinzhou Women and Children's Hospital, Jinzhou, China
| | - Xuezhe Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Kun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
35
|
Li X, Shen B, Feng F, Li K, Tang Z, Ma L, Li H. Dual-view jointly learning improves personalized drug synergy prediction. Bioinformatics 2024; 40:btae604. [PMID: 39423102 PMCID: PMC11524890 DOI: 10.1093/bioinformatics/btae604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/23/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024] Open
Abstract
MOTIVATION Accurate and robust estimation of the synergistic drug combination is important for medicine precision. Although some computational methods have been developed, some predictions are still unreliable especially for the cross-dataset predictions, due to the complex mechanism of drug combinations and heterogeneity of cancer samples. RESULTS We have proposed JointSyn that utilizes dual-view jointly learning to predict sample-specific effects of drug combination from drug and cell features. JointSyn outperforms existing state-of-the-art methods in predictive accuracy and robustness across various benchmarks. Each view of JointSyn captures drug synergy-related characteristics and makes complementary contributes to the final prediction of the drug combination. Moreover, JointSyn with fine-tuning improves its generalization ability to predict a novel drug combination or cancer sample using a small number of experimental measurements. We also used JointSyn to generate an estimated atlas of drug synergy for pan-cancer and explored the differential pattern among cancers. These results demonstrate the potential of JointSyn to predict drug synergy, supporting the development of personalized combinatorial therapies. AVAILABILITY AND IMPLEMENTATION Source code and data are available at https://github.com/LiHongCSBLab/JointSyn.
Collapse
Affiliation(s)
- Xueliang Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bihan Shen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fangyoumin Feng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kunshi Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhixuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liangxiao Ma
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Science, Shanghai 200031, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
36
|
Mahmoudi A, Jamialahmadi T, Kesharwani P, Sahebkar A. Bioinformatic analysis of the molecular targets of curcumin in colorectal cancer. Pathol Res Pract 2024; 262:155533. [PMID: 39173464 DOI: 10.1016/j.prp.2024.155533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024]
Abstract
Colorectal cancer (CRC) is a major global health concern, with rising incidence and mortality rates. Conventional treatments often come with significant complications, prompting the exploration of natural compounds like curcumin as potential therapeutic agents. Using bioinformatic tools, this study investigated the role of curcumin in CRC treatment. Significant protein interactions between curcumin and target proteins were identified in the STITCH database. Differentially expressed genes (DEGs) associated with CRC were then analyzed from GEO databases. Comparing curcumin targets and CRC-related DEGs, nine significant common targets were identified: DNMT1, PCNA, CCND1, PLAU, MMP3, SOX9, FOXM1, CXCL2, and SERPINB5. Pathway enrichment analyses revealed that curcumin-targeted pathways were primarily related to p53, IL-17, NF-kappa B, TNF, and cell cycle signaling, all crucial in CRC development and progression. Further analyses using DAID and EnrichR algorithms showed that the curcumin targets exhibited greater specificity to bronchial epithelial cells and colorectal adenocarcinoma than other diseases. Analyses via the DSigDB database indicated that curcumin ranks highly among other drugs targeting the identified CRC-related genes. Docking studies revealed favorable binding interactions between curcumin and the key CRC-related proteins, suggesting potential molecular mechanisms by which curcumin may exert its effects. In summary, this study provides bioinformatic and docking evidence that curcumin may exert beneficial effects on CRC by modulating the expression or activity of multiple CRC-susceptibility genes involved in critical signaling pathways. These findings warrant further experimental validation and support the potential of curcumin as a therapeutic agent for CRC.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Yang Y, Hao X, Zhang J, Gao T, Huo M, Liu W, Hu T, Ma T, Yuan B, Zhang M, Teng X, Yu H, Huang W, Wang Y. The E3 ligase TRIM22 functions as a tumor suppressor in breast cancer by targeting CCS for proteasomal degradation to inhibit STAT3 signaling. Cancer Lett 2024; 600:217157. [PMID: 39127340 DOI: 10.1016/j.canlet.2024.217157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/02/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Deregulation of E3 ubiquitin ligases drives the proliferation and metastasis of various cancers; however, the underlying mechanisms remain unknown. This study aimed to investigate the role of tripartite motif-containing 22 (TRIM22), a poorly investigated E3 ubiquitin ligase in the TRIM family, as a tumor suppressor in breast cancer. High expression of TRIM22 in breast cancer correlated with better prognosis. Functional experiments demonstrated that TRIM22 significantly inhibited the proliferation and invasion of breast cancer cells. Label-free proteomics and biochemical analyses revealed that the copper chaperone for superoxide dismutase (CCS), an oncoprotein that is upregulated in breast cancer and promotes the growth and invasion of breast cancer cells, was a target of TRIM22 for degradation via K27-linked ubiquitination. Notably, the ability of the coiled-coil domain-defective mutants of TRIM22 to induce CCS ubiquitination and degradation diminished, with lysine 76 of the CCS serving as the ubiquitination site. Moreover, the TRIM22-mediated inhibition of the proliferation and invasion of breast cancer cells was restored by ectopic CCS expression. RNA-sequencing experiments using Gene Set Enrichment Analysis demonstrated that TRIM22 is involved in the JAK-STAT signaling pathway. TRIM22 overexpression also improved reactive oxygen species levels in breast cancer cells and inhibited STAT3 phosphorylation, which was restored via CCS overexpression or N-acetyl-l-cysteine treatment. Chromatin immunoprecipitation-quantitative polymerase chain reaction results showed that TRIM22 overexpression decreased the enrichment of phosphorylated STAT3 in FN1, VIM and JARID2 promoters. Clinically, low TRIM22 expression correlated with high CCS expression and decreased survival rates in patients with breast cancer. Moreover, TRIM22 upregulation was associated with a better prognosis in patients with breast cancer who received classical therapy. TRIM22 expression was downregulated in many cancer types, including colon, kidney, lung, and prostate cancers. To the best of our knowledge, the E3 ubiquitin ligase TRIM22 was first reported as a tumor suppressor that inhibits the proliferation and invasion of breast cancer cells through CCS ubiquitination and degradation. TRIM22 is a potential prognostic biomarker in patients with breast cancer.
Collapse
Affiliation(s)
- Yunkai Yang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinhui Hao
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jingyao Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianyang Gao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Miaomiao Huo
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wei Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ting Hu
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianyu Ma
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Baowen Yuan
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Min Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xu Teng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hefen Yu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Sciences, Zhengzhou, Henan Province, 450000, China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
38
|
Chen Y, Yang S, Yu T, Zeng T, Wei L, You Y, Tang J, Dang T, Sun H, Zhang Y. KDM4A promotes malignant progression of breast cancer by down-regulating BMP9 inducing consequent enhancement of glutamine metabolism. Cancer Cell Int 2024; 24:322. [PMID: 39300582 DOI: 10.1186/s12935-024-03504-0] [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: 07/21/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Recent studies have found that histone-modified genes play an increasingly important role in tumor progression. Lysine(K) specific demethylase 4A (KDM4A) is a histone lysine-specific demethylase highly expressed in a variety of malignant tumors, data showed that KDM4A was negatively correlated with the Bone Morphogenetic Protein 9 (BMP9) in breast cancer. And previous experiments have demonstrated that exogenous BMP9 significantly inhibits breast cancer development. MATERIALS AND METHODS We detected the expression of KDM4A in breast cancer and the relationship between KDM4A and BMP9 using real-time quantitative PCR (RT-qPCR) and Western blot, and verified the interaction between KDM4A and BMP9 by ChIP experiments. At the same time, we also detected whether KDM4A had effects on the RNA and protein stability of BMP9 using actinomycin D and cycloheximide. Measurement of alpha-ketoglutarate (α-KG) level by ELISA to observe the effect of BMP9 on glutamine metabolism in breast cancer cells. Nucleoplasmic distribution of KDM4A after exogenous BMP9 treatment in breast cancer cells were observed by immunofluorescence staining and Western blot. A subcutaneous xenograft tumor model in nude mice was used to study the therapeutic effects of exogenous BMP9 and KDM4A inhibitor (JIB-04) in breast cancer. CCK-8, conoly formation, Transwell, wound healing, and immunohistochemistry were used to monitor the growth of tumor and cell function. RESULTS We found that KDM4A was abnormally highly expressed in breast cancer, and silenced BMP9 expression by removing histone methyl groups from the BMP9 gene region. Meanwhile, KDM4A could also reduce the stability of BMP9 protein. BMP9 inhibit glutamine metabolism in breast cancer, resulting in a decrease in its product α-KG, is confirmed by ELISA. Altered nucleoplasmic distribution of KDM4A due to decreased α-KG was confirmed by immunofluorescence staining and Western blot. Animal experiments confirm that the combination of exogenous BMP9 and JIB-04 shows significantly better results in breast cancer. CONCLUSIONS KDM4A silences BMP9 expression by removing histone methyl groups from the BMP9 gene region, leading to further enhancement of glutamine metabolism, which contributes to malignant tumor progression. In addition, using JIB-04 in combination with exogenous BMP9 could inhibit the malignant progression of breast cancer cells and the growth of tumors more significantly.
Collapse
Affiliation(s)
- Yuanxiang Chen
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shiyu Yang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tao Yu
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tao Zeng
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lan Wei
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yiqing You
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jiafeng Tang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tingting Dang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Haoli Sun
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yan Zhang
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
39
|
Dhiman R, Bazad N, Mukherjee R, Himanshu, Gunjan, Leal E, Ahmad S, Kaur K, Raj VS, Chang CM, Pandey RP. Enhanced drug delivery with nanocarriers: a comprehensive review of recent advances in breast cancer detection and treatment. DISCOVER NANO 2024; 19:143. [PMID: 39243326 PMCID: PMC11380656 DOI: 10.1186/s11671-024-04086-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer (BC) remains a leading cause of morbidity and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges due to its aggressive phenotype and resistance to conventional therapies. Recent advancements in nanocarrier technology offer promising solutions for enhancing drug delivery, improving bioavailability, and increasing drug accumulation at tumor sites through targeted approaches. This review delves into the latest innovations in BC detection and treatment, highlighting the role of nanocarriers like polymeric micelles, liposomes, and magnetic nanoparticles in overcoming the limitations of traditional therapies. Additionally, the manuscript discusses the integration of cutting-edge diagnostic tools, such as multiplex PCR-Nested Next-Generation Sequencing (mPCR-NGS) and blood-based biomarkers, which are revolutionizing early detection and molecular profiling of BC. The convergence of these technologies not only enhances therapeutic outcomes but also paves the way for personalized medicine in BC management. This comprehensive review underscores the potential of nanocarriers in transforming BC treatment and emphasizes the critical importance of early detection in improving patient prognosis.
Collapse
Affiliation(s)
- Ruby Dhiman
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
- School of Health Sciences and Technology (SOHST), UPES, Dehradun, Uttarakhand, 248007, India
| | - Nancy Bazad
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Riya Mukherjee
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Himanshu
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Gunjan
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Elcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belem, Pará, Brazil
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail City, Kingdom of Saudi Arabia
| | - Kirtanjot Kaur
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Chung-Ming Chang
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC.
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan City, Taiwan, ROC.
- Laboratory Animal Center, Chang Gung University, Taoyuan City, Taiwan, ROC.
| | - Ramendra Pati Pandey
- School of Health Sciences and Technology (SOHST), UPES, Dehradun, Uttarakhand, 248007, India.
| |
Collapse
|
40
|
Heydarnia E, Sepasi A, Asefi N, Khakshournia S, Mohammadnejad J. The effects of metformin and PCL-sorafenib nanoparticle co-treatment on MCF-7 cell culture model of breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7213-7221. [PMID: 38656346 DOI: 10.1007/s00210-024-03049-z] [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: 10/28/2023] [Accepted: 03/10/2024] [Indexed: 04/26/2024]
Abstract
Despite breakthrough therapeutics in breast cancer, it is one of the main causes of mortality among women worldwide. Thus, drug therapies for treating breast cancer have recently been developed by scientists. Metformin and sorafenib are well-known therapeutics in breast cancer. In the present study, we combined sorafenib and PCL-sorafenib with metformin to improve drug absorption and promote therapeutic efficiency. The MCF-7 cells were treated with metformin, sorafenib, or PCL-sorafenib. The growth inhibitory effect of these drugs and cell viability were assessed using MTT and flow cytometry assays, respectively. The expression of targeted genes involved in cell proliferation, signaling, and the cell cycle was measured by real-time PCR. The results showed that MCF-7 cells treated with metformin/sorafenib and PCL-sorafenib/metformin co-treatment contributed to 50% viability compared to the untreated group. Moreover, PI and Annexin V staining tests showed that the cell viability for metformin/sorafenib and PCL-sorafenib/metformin was 38% and 17%, respectively. Furthermore, sorafenib/metformin and PCL-sorafenib/metformin lead to p53 gene expression increase by which they can increase ROS, thereby decreasing GPX4 gene expression. In addition, they affected the expression of BCL2 and BAX genes and altered the cell cycle. Together, the combination of PCL-sorafenib/metformin and sorafenib/metformin increased sorafenib absorption at lower doses and also led to apoptosis and oxidative stress increases in MCF-7 cells.
Collapse
Affiliation(s)
- Emad Heydarnia
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aref Sepasi
- Department of Medical Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, Tehran, Iran
| | - Nika Asefi
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, Tehran, Iran
| | - Sara Khakshournia
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, 14395-1561, Iran.
| |
Collapse
|
41
|
Abumansour H, Abusara OH, Khalil W, Abul-Futouh H, Ibrahim AIM, Harb MK, Abulebdah DH, Ismail WH. Biological evaluation of levofloxacin and its thionated derivatives: antioxidant activity, aldehyde dehydrogenase enzyme inhibition, and cytotoxicity on A549 cell line. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6963-6973. [PMID: 38613572 DOI: 10.1007/s00210-024-03075-x] [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: 03/09/2024] [Accepted: 03/28/2024] [Indexed: 04/15/2024]
Abstract
Levofloxacin (LVX) is among the fluoroquinolones antibiotics that has also been studied in vitro and in vivo for its anticancer effects. In this study, we used LVX and novel LVX thionated derivatives; compounds 2 and 3, to evaluate their antioxidant activity, aldehyde dehydrogenase (ALDH) enzymes activity inhibition, and anticancer activity. Combination treatments with doxorubicin (DOX) were investigated as well. The 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay was used to determine the antioxidant activity. The NADH fluorescence spectrophotometric activity assay was used to determine the ALDH inhibitory effects. Resazurin dye method was applied for cell viability assays. Molecular Operating Environment software was used for the molecular docking experiments. Compared to ascorbic acid, DPPH assay showed that compound 3 had the highest antioxidant activity among the tested compounds with approximately 35% scavenging activity. On ALDH enzymes, compound 3 showed a significant ALDH activity inhibition compared to compound 2 at 200 µM. The IC50 values for the tested compounds were approximately 100 µM on A549 cell line, a non-small cell lung cancer (NSCLC) cell line. However, significant enhancement of cytotoxicity and reduction of IC50 values were observed by combining DOX and synergism was achieved with LVX with a combination index value of 0.4. The molecular docking test showed a minimum binding energy with a good affinity for compound 3 towards ALDH enzymes. Thionated LVX derivatives, may be repurposed for NSCLC therapy in combination with DOX, taking into account the antioxidant activity, ALDH activity inhibition, and the molecular docking results of compound 3.
Collapse
Affiliation(s)
- Hamza Abumansour
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan.
| | - Osama H Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Wiam Khalil
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Hassan Abul-Futouh
- Department of Chemistry, Faculty of Science, The Hashemite University, P.O. Box 330127, Zarqa, 13133, Jordan
| | - Ali I M Ibrahim
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Mohammad K Harb
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Dina H Abulebdah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| | - Worood H Ismail
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman, 11733, Jordan
| |
Collapse
|
42
|
Chen X, Ding X, Fang J, Mao C, Gong X, Zhang Y, Zhang N, Yan F, Lou Y, Chen Z, Ding W, Ma Z. Natural Derivatives of Selective HDAC8 Inhibitors with Potent in Vivo Antitumor Efficacy against Breast Cancer. J Med Chem 2024; 67:14609-14632. [PMID: 39110628 DOI: 10.1021/acs.jmedchem.4c01438] [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: 08/23/2024]
Abstract
HDAC8 is a therapeutic target with great promise for breast cancer. Here, we reported a novel compound corallorazine D from Nocardiopsis sp. XZB108, selectively inhibited HDAC8 (IC50 = 0.90 ± 0.014 μM), suggesting that it may be a promising nonhydroxamate HDAC8 inhibitor. Upon additional modifications of corallorazine D, a candidate compound 5k, demonstrated remarkable inhibitory potency against HDAC8 (IC50 = 0.12 ± 0.01 nM), 89-fold superior to PCI-34051. The selectivity of 5k was at least 439-fold, superior to corallorazine D, confirming the efficacy of our modifications. In an orthotopic mouse model of breast cancer, 5k displayed nearly 4-fold superior antitumor activity than SAHA. Furthermore, 5k triggered antitumor immunity by activating T cells. Treatment with 5k significantly increased the proportion of M1 macrophages and decreased the proportion of M2 macrophages (M1/M2 ratio = 2.67 ± 0.25). 5k represents a promising compound for further investigation as a potential treatment for breast cancer.
Collapse
Affiliation(s)
- Xiaoming Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xia Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Jiebin Fang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Churu Mao
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xingzhi Gong
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yuxiao Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Ningjing Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Feihang Yan
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yijie Lou
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wanjing Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Zhongjun Ma
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| |
Collapse
|
43
|
Wang W, Huang S, Li S, Li X, Ling Y, Wang X, Zhang S, Zhou D, Yin W. Rosa sterilis Juice Alleviated Breast Cancer by Triggering the Mitochondrial Apoptosis Pathway and Suppressing the Jak2/Stat3 Pathway. Nutrients 2024; 16:2784. [PMID: 39203920 PMCID: PMC11357216 DOI: 10.3390/nu16162784] [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: 07/09/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Rosa sterilis (RS) is a characteristic fruit in southwestern China that has numerous health benefits; however, its pharmacological effect needs further clarification, especially with respect to the exploration of its potential anti-breast-cancer effect, as there are still knowledge gaps in this regard. This study was designed to investigate the protective effects of Rosa sterilis juice (RSJ) on breast cancer (BC) through in vitro cellular experiments and by establishing mouse 4T1 breast xenograft tumors. This study also had the aim of elucidating RSJ's underlying mechanisms. RSJ can inhibit cell proliferation, affect cell morphology, and impact the clone formation ability of BC; furthermore, it can promote apoptosis by triggering the mitochondrial apoptosis pathway. In mouse 4T1 breast xenograft tumors, RSJ markedly inhibited tumor growth, relieved the pathological lesions, lowered the expression of Ki67, and regulated the expression of the apoptosis-associated protein. Moreover, we observed that RSJ can inhibit the Jak2/Stat3 signaling pathway both in vivo and in vitro. Overall, our research reveals that RSJ can alleviate BC by triggering the mitochondrial apoptosis pathway and suppressing the Jak2/Stat3 pathway, providing new dietary intervention strategies for BC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dingzi Zhou
- West China School of Public Health and West China Fourth Hospital, Sichuan University, 17# 3rd Section, Ren Min South Road, Chengdu 610041, China; (W.W.); (S.H.); (S.L.); (X.L.); (Y.L.); (X.W.); (S.Z.)
| | - Wenya Yin
- West China School of Public Health and West China Fourth Hospital, Sichuan University, 17# 3rd Section, Ren Min South Road, Chengdu 610041, China; (W.W.); (S.H.); (S.L.); (X.L.); (Y.L.); (X.W.); (S.Z.)
| |
Collapse
|
44
|
Winter RC, Amghar M, Wacker AS, Bakos G, Taş H, Roscher M, Kelly JM, Benešová-Schäfer M. Future Treatment Strategies for Cancer Patients Combining Targeted Alpha Therapy with Pillars of Cancer Treatment: External Beam Radiation Therapy, Checkpoint Inhibition Immunotherapy, Cytostatic Chemotherapy, and Brachytherapy. Pharmaceuticals (Basel) 2024; 17:1031. [PMID: 39204136 PMCID: PMC11359268 DOI: 10.3390/ph17081031] [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: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer is one of the most complex and challenging human diseases, with rising incidences and cancer-related deaths despite improved diagnosis and personalized treatment options. Targeted alpha therapy (TαT) offers an exciting strategy emerging for cancer treatment which has proven effective even in patients with advanced metastatic disease that has become resistant to other treatments. Yet, in many cases, more sophisticated strategies are needed to stall disease progression and overcome resistance to TαT. The combination of two or more therapies which have historically been used as stand-alone treatments is an approach that has been pursued in recent years. This review aims to provide an overview on TαT and the four main pillars of therapeutic strategies in cancer management, namely external beam radiation therapy (EBRT), immunotherapy with checkpoint inhibitors (ICI), cytostatic chemotherapy (CCT), and brachytherapy (BT), and to discuss their potential use in combination with TαT. A brief description of each therapy is followed by a review of known biological aspects and state-of-the-art treatment practices. The emphasis, however, is given to the motivation for combination with TαT as well as the pre-clinical and clinical studies conducted to date.
Collapse
Affiliation(s)
- Ruth Christine Winter
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mariam Amghar
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Anja S. Wacker
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Gábor Bakos
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Harun Taş
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mareike Roscher
- Service Unit for Radiopharmaceuticals and Preclinical Studies, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - James M. Kelly
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| |
Collapse
|
45
|
Lessa VL, Gonçalves G, Santos B, Cavalari VC, da Costa Vieira RF, Figueiredo FB. In Vitro Evaluation of the Combinatorial Effect of Naringenin and Miltefosine against Leishmania amazonensis. Pharmaceuticals (Basel) 2024; 17:1014. [PMID: 39204118 PMCID: PMC11357177 DOI: 10.3390/ph17081014] [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: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Leishmania amazonensis causes a clinical form called diffuse cutaneous leishmaniasis (DCL) with challenges to treatment, like low efficiency and drug toxicity. Therefore, it is necessary to investigate new therapies using less toxic leishmanicidal compounds, such as flavonoids like naringenin, and their combination with conventional drugs, such as miltefosine. Antileishmanial dose/response activity, isobologram, calculation of dose reduction index (DRI), and fractional inhibitory concentration index (FICI) tests were performed on in vitro assays using reference promastigote forms of L. amazonensis (IFLA/BR/67/PH8) to assess the combinatorial effect between naringenin and miltefosine. The in vitro results of isobologram, DRI, and FICI calculations showed that the combination of the compounds had an additive effect and was able to reduce the half maximal inhibitory concentration (IC50) of miltefosine in the promastigote forms of the parasite compared to the treatment of the drug alone. This study demonstrated in vitro the viability of a combination action of the flavonoid with the treatment with miltefosine, opening space for further investigations on the association of natural compounds with the drugs used for the treatment of L. amazonensis.
Collapse
Affiliation(s)
- Vinícius Lopes Lessa
- Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81310-020, PR, Brazil; (G.G.); (B.S.); (V.C.C.); (F.B.F.)
- Graduate Program in Veterinary Sciences, Federal University of Paraná, Curitiba 80035-060, PR, Brazil
| | - Gustavo Gonçalves
- Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81310-020, PR, Brazil; (G.G.); (B.S.); (V.C.C.); (F.B.F.)
| | - Beatriz Santos
- Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81310-020, PR, Brazil; (G.G.); (B.S.); (V.C.C.); (F.B.F.)
| | - Victoria Cruz Cavalari
- Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81310-020, PR, Brazil; (G.G.); (B.S.); (V.C.C.); (F.B.F.)
| | - Rafael Felipe da Costa Vieira
- Department of Epidemiology and Community Health, College of Health and Human Services, University of North Carolina at Charlotte, Charlotte, NC 28223, USA;
- Center for Computational Intelligence to Predict Health and Environmental Risks (CIPHER), University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Fabiano Borges Figueiredo
- Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81310-020, PR, Brazil; (G.G.); (B.S.); (V.C.C.); (F.B.F.)
| |
Collapse
|
46
|
Du Y, Cao Y, Song W, Wang X, Yu Q, Peng X, Zhao R. Role of the P2X7 receptor in breast cancer progression. Purinergic Signal 2024:10.1007/s11302-024-10039-6. [PMID: 39039304 DOI: 10.1007/s11302-024-10039-6] [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: 03/23/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
Breast cancer is a common malignant tumor, whose incidence is increasing year by year, and it has become the malignant tumor with the highest incidence rate in women. Purine ligand-gated ion channel 7 receptor (P2X7R) is a cation channel receptor with Adenosine triphosphate ( ATP) as a ligand, which is widely distributed in cells and tissues, and is closely related to tumorigenesis and progression. P2X7R plays an important role in cancer by interacting with ATP. Studies have shown that P2X7R is up-regulated in breast cancer and can promote tumor invasion and metastasis by activating the protein kinase B (AKT) signaling pathway, promoting epithelial-mesenchymal transition (EMT), controlling the generation of extracellular vesicle (EV), and regulating the expression of the inflammatory protein cyclooxygenase 2 (COX-2). Furthermore, P2X7R was proven to play an essential role in the proliferation and apoptosis of breast cancer cells. Recently, inhibitors targeting P2X7R have been found to inhibit the progression of breast cancer. Natural P2X7R antagonists, such as rhodopsin, and the isoquinoline alkaloid berberine, have also been shown to be effective in inhibiting breast cancer progression. In this article, we review the research progress of P2X7R and breast cancer intending to provide new targets and directions for breast cancer treatment.
Collapse
Affiliation(s)
- Yanan Du
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Yahui Cao
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Wei Song
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Xin Wang
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Qingqing Yu
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China
| | - Xiaoxiang Peng
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China.
| | - Ronglan Zhao
- School of Medical Laboratory, Shandong Second Medical University, Shandong, 261053, China.
| |
Collapse
|
47
|
Kaur S, Mendonca P, Soliman KFA. The Anticancer Effects and Therapeutic Potential of Kaempferol in Triple-Negative Breast Cancer. Nutrients 2024; 16:2392. [PMID: 39125273 PMCID: PMC11314279 DOI: 10.3390/nu16152392] [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: 06/15/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer is the second-leading cause of cancer death among women in the United States. Triple-negative breast cancer (TNBC), a subtype of breast cancer, is an aggressive phenotype that lacks estrogen (ER), progesterone (PR), and human epidermal growth (HER-2) receptors, which is challenging to treat with standardized hormonal therapy. Kaempferol is a natural flavonoid with antioxidant, anti-inflammatory, neuroprotective, and anticancer effects. Besides anti-tumorigenic, antiproliferative, and apoptotic effects, kaempferol protects non-cancerous cells. Kaempferol showed anti-breast cancer effects by inducing DNA damage and increasing caspase 3, caspase 9, and pAMT expression, modifying ROS production by Nrf2 modulation, inducing apoptosis by increasing cleaved PARP and Bax and downregulating Bcl-2 expression, inducing cell cycle arrest at the G2/M phase; inhibiting immune evasion by modulating the JAK-STAT3 pathway; and inhibiting the angiogenic and metastatic potential of tumors by downregulating MMP-3 and MMP-9 levels. Kaempferol holds promise for boosting the efficacy of anticancer agents, complementing their effects, or reversing developed chemoresistance. Exploring novel TNBC molecular targets with kaempferol could elucidate its mechanisms and identify strategies to overcome limitations for clinical application. This review summarizes the latest research on kaempferol's potential as an anti-TNBC agent, highlighting promising but underexplored molecular pathways and delivery challenges that warrant further investigation to achieve successful clinical translation.
Collapse
Affiliation(s)
- Sukhmandeep Kaur
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| |
Collapse
|
48
|
Wang YK, Zhu RW, Gao ZP, Tao Y. Quality of life and survival analyses of breast cancer cases treated with integrated traditional Chinese and Western medicine. World J Clin Cases 2024; 12:4074-4081. [PMID: 39015919 PMCID: PMC11235521 DOI: 10.12998/wjcc.v12.i20.4074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the second leading cause of tumor-related mortality after lung cancer. Chemotherapy resistance remains a major challenge to progress in BC treatment, warranting further exploration of feasible and effective alternative therapies. AIM To analyzed the quality of life (QoL) and survival of patients with BC treated with integrated traditional Chinese and Western medicine (TCM-WM). METHODS This study included 226 patients with BC admitted to the First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine between February 2018 and February 2023, including 100 who received conventional Western medicine treatment (control group) and 126 who received TCM-WM treatment (research group). The total effective rate, side effects (alopecia, nausea and vomiting, hepatorenal toxicity, and myelosuppression), QoL assessed using the European Organization for Research and Treatment of Cancer Core Quality of Life Questionnaire (EORTC QLQ-C30), 1-year overall survival (OS), recurrence and metastasis rates, and serum inflammatory factors [interleukin (IL)-6, IL-10, and tumor necrosis factor alpha] were comparatively analyzed. RESULTS The research group showed statistically better overall efficacy, EORTC QoL-C30 scores, and 1-year OS than the control group, with markedly lower side effects and 1-year recurrence and metastasis rates. Moreover, the posttreatment levels of serum inflammatory in the research group were significantly lower than the baseline and those in the control group. CONCLUSION Overall, TCM-WM demonstrated significantly improved therapeutic efficacy while ensuring drug safety in BC, which not only improved patients' QoL and prolonged survival, but also significantly inhibited the inflammatory response.
Collapse
Affiliation(s)
- Yu-Kun Wang
- Thyroid and Breast Surgery Department, the First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Rong-Wei Zhu
- Clinical Integration of Traditional Chinese and Western Medicine, Guangxi University of Chinese Medicine, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Peng Gao
- Clinical Integration of Traditional Chinese and Western Medicine, Guangxi University of Chinese Medicine, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Yi Tao
- Clinical Integration of Traditional Chinese and Western Medicine, Guangxi University of Chinese Medicine, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
49
|
Passos CLA, Ferreira C, de Carvalho AGA, Silva JL, Garrett R, Fialho E. Oxyresveratrol in Breast Cancer Cells: Synergistic Effect with Chemotherapeutics Doxorubicin or Melphalan on Proliferation, Cell Cycle Arrest, and Cell Death. Pharmaceutics 2024; 16:873. [PMID: 39065570 PMCID: PMC11279446 DOI: 10.3390/pharmaceutics16070873] [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: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the second most common type of cancer in the world. Polyphenols can act at all stages of carcinogenesis and oxyresveratrol (OXY) promising anticancer properties, mainly associated with chemotherapy drugs. The aim of this study was to investigate the effect of OXY with doxorubicin (DOX) or melphalan (MEL), either isolated or associated, in MCF-7 and MDA-MB-231 breast cancer cells. Our results showed that OXY, DOX, and MEL presented cytotoxicity, in addition to altering cell morphology. The synergistic association of OXY + DOX and OXY + MEL reduced the cell viability in a dose-dependent manner. The OXY, DOX, or MEL and associations were able to alter the ROS production, ∆Ψm, and cell cycle; DOX and OXY + DOX led the cells to necrosis. Furthermore, OXY and OXY + MEL were able to lead the cells to apoptosis and upregulate caspases-3, -7, -8, and -9 in both cells. LC-HRMS showed that 7-deoxidoxorubicinone and doxorubicinol, responsible for the cardiotoxic effect, were not identified in cells treated with the OXY + DOX association. In summary, our results demonstrate for the first time the synergistic effect of OXY with chemotherapeutic agents in breast cancer cells, offering a new strategy for future animal studies.
Collapse
Affiliation(s)
- Carlos Luan Alves Passos
- Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.L.A.P.); (C.F.)
| | - Christian Ferreira
- Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.L.A.P.); (C.F.)
| | | | - Jerson Lima Silva
- Medical Biochemistry Institute Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Rafael Garrett
- Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, RJ, Brazil; (A.G.A.d.C.); (R.G.)
| | - Eliane Fialho
- Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.L.A.P.); (C.F.)
| |
Collapse
|
50
|
Erden Tayhan S. A study with cancer stem cells and three-dimensional tumoroids: investigation of the combined effects of 5-fluorouracil and doxorubicin in breast cancer. Med Oncol 2024; 41:185. [PMID: 38910198 PMCID: PMC11194218 DOI: 10.1007/s12032-024-02423-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
The purpose of the present study was in vitro determination of the combined effects of doxorubucin and 5-fluorouracil by 2D and 3D culture conditions on breast cancer using MCF-7 cell line and CSCs isolated from these cells. In the first stage of this study, CSC isolation and their characterization were performed. In the next experimental period, the antiproliferative effects of 5-Fu and Dox on the MCF-7 and CSCs were demonstrated on 2D. To evaluate the synergistic/antagonistic effects of these chemotherapeutics, the CI was calculated. Additionally, 3D tumor spheroids were used as another model. In the last step, qRT-PCR analysis was performed to examine apoptosis-related gene expressions. In this study, it was clearly seen that CSCs obtained from the breast cancer cell line express stemness factors. In addition, the antiproliferative effects of 5-Fu and Dox on breast cancer and associated CSCs were very clear. Their synergistic effects were determined by CI values. Moreover, it was seen that combined theraphy changed the expression levels of genes related to apoptosis. Additionally, it was molecularly demonstrated that 3D tumoroids were more resistant than the others. In conclusion, the polychemotherapeutic approach was much more effective than the monotherapy. The fact that this effect was seen not only in breast cancer cells, but also in breast cancer stem cells. In addition, it was very promising that the results obtained were similar in both two-dimensional and three-dimensional tumoroids.
Collapse
Affiliation(s)
- Seçil Erden Tayhan
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|