1
|
Jiang M, Zhang K, Meng J, Xu L, Liu Y, Wei R. Engineered exosomes in service of tumor immunotherapy: From optimizing tumor-derived exosomes to delivering CRISPR/Cas9 system. Int J Cancer 2025; 156:898-913. [PMID: 39474936 DOI: 10.1002/ijc.35241] [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: 07/17/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 01/07/2025]
Abstract
Exosomes can be modified and designed for various therapeutic goals because of their unique physical and chemical characteristics. Researchers have identified tumor-derived exosomes (TEXs) as significant players in cancer by influencing tumor growth, immune response evasion, angiogeneis, and drug resistance. TEXs promote the production of specific proteins important for cancer progression. Due to their easy accessibility, TEXs are being modified through genetic, drug delivery, membrane, immune system, and chemical alterations to be repurposed as vehicles for delivering drugs to improve cancer treatment outcomes. In the complex in vivo environment, the clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9) system encounters challenges from degradation, neutralization, and immune responses, emphasizing the need for strategic distribution strategies for effective genome editing. Engineered exosomes present a promising avenue for delivering CRISPR/Cas9 in vivo. In this review, we will explore different techniques for enhancing TEXs using various engineering strategies. Additionally, we will discuss how these exosomes can be incorporated into advanced genetic engineering systems like CRISPR/Cas9 for possible therapeutic uses.
Collapse
Affiliation(s)
- Mingyang Jiang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ke Zhang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinfeng Meng
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Linhua Xu
- The First Clinical Medical College, Guangxi Medical University, Nanning
| | - Ying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruqiong Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Ma S, Jiang L, Yang W, Liu F, Wang D, Wang F, Huang J. Advances of Nanomaterials in Cancer Photocatalysis Therapy. MATERIALS TODAY SUSTAINABILITY 2025; 29:101023. [DOI: 10.1016/j.mtsust.2024.101023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Abdul-Razek N, Khalil RG, Abdel-Latif M, Kamel MM, Alhazza IM, Awad EM, Ebaid H, Abuelsaad ASA. Investigating the Tumor-Suppressive, Antioxidant Effects and Molecular Binding Affinity of Quercetin-Loaded Selenium Nanoparticles in Breast Cancer Cells. BIONANOSCIENCE 2025; 15:135. [DOI: 10.1007/s12668-024-01767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/03/2025]
Abstract
AbstractIn 2023, breast cancer is expected to have nearly 2 million new cases, making it the second most common cancer overall and the most prevalent among women. Multidrug resistance limits the effectiveness of chemotherapy; however, quercetin, a natural flavonoid, helps combat this issue. The goal of the current investigation is to determine the impact of a novel composite of quercetin and selenium nanoparticles (SeNPs) on the breast cancer cell lines MDA-MB-231 and MCF-7 in order to enhance quercetin’s tumor-suppressive action and decrease selenium (Se) toxicity. Particle size, zeta potential, FTIR, SEM, UV–VIS spectroscopy, and EDX were used to characterize quercetin-selenium nanoparticles (Que-SeNPs), in addition to evaluation of the antioxidant, apoptotic, and anticancer properties. Moreover, autophagy (Atg-13) protein receptors and PD-1/PD-L1 checkpoint were targeted using molecular docking modeling and molecular dynamics (MD) simulations to assess the interaction stability between Que-SeNPs and three targets: PDL-1, PD-1, and Atg-13HORMA domain. Que-SeNPs, synthesized with quercetin, were stable, semi-spherical (80–117 nm), and had a zeta potential of − 37.8 mV. They enhanced cytotoxicity, antioxidant activity, and apoptosis compared to quercetin alone in MCF-7 and MDA-MB-231 cells. Docking simulations showed strong binding to the PD-1/PD-L1 checkpoint and Atg-13HORMA protein receptors. Moreover, the molecular dynamics simulation revealed that the behavior of the PD-L1 intriguing insights into its structural dynamics, therefore, suggesting a stable phase where the complex is adjusting to the simulation environment. The present data confirmed that the stable formula of Que-SeNPs is cytotoxic, antioxidant, and has a potential activity to increase apoptosis in breast cancer cells, with the potential to inhibit PD-1/PD-L1 and Atg-13 proteins.
Graphical Abstract
Role of Que-SeNPs on breast cancer cells in vitro against two breast cancer cell lines MDA-MB-231 and MCF-7.
Collapse
|
4
|
Roman M, Wrobel TP, Panek A, Kwiatek WM. Comparison of biochemical changes induced in radioresistant prostate cancer cells by X-rays, radiosensitizing drugs, and a combined therapy using Raman microspectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 326:125218. [PMID: 39353252 DOI: 10.1016/j.saa.2024.125218] [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: 01/31/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Cancer radioresistance is a major problem in radiotherapy. Many strategies have been proposed to overcome this process including the use of radiosensitizing drugs such as C75 or silibinin. The overall result of all treatments (radiotherapy, chemotherapy, and combined treatment) is cancer cell death. On the other hand, each treatment affects cancer cells differently at the molecular level. However, little is known about biochemical changes induced in cancer cells by these treatments (especially in combined therapy) at the submicroscale. In this study, Raman microspectroscopy was applied to follow such changes induced in radioresistant prostate cancer cells by X-rays, radiosensitizing drugs (C75, silibinin), and a combined treatment. The analysis was supported by the Partial Least Squares Regression method to reveal spectral changes induced by an increasing dose of X-rays and concentrations of the drugs. The obtained regression coefficient (β) plots were compared to each other using a correlation coefficient (R). Our results show that PC-3 cells exhibit dose- and concentration-dependent responses to the treatment with different biochemical changes induced by X-rays in the presence of C75 and silibinin. Moreover, both drugs affect the cells differently at the submicroscale and independently from the X-ray's presence. Finally, C75 shows significant efficiency in the reduction of cell radioresistance.
Collapse
Affiliation(s)
- Maciej Roman
- Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland; SOLARIS National Synchrotron Radiation Centre, Jagiellonian University, Czerwone Maki 98, 30-392 Krakow, Poland.
| | - Tomasz P Wrobel
- SOLARIS National Synchrotron Radiation Centre, Jagiellonian University, Czerwone Maki 98, 30-392 Krakow, Poland
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| | - Wojciech M Kwiatek
- Institute of Nuclear Physics Polish Academy of Sciences, Radzikowskiego 152, 31-342 Krakow, Poland
| |
Collapse
|
5
|
Tufail T, Ain HBU, Virk MS, Ashraf J, Ahmed Z, Khalil AA, Rasheed A, Xu B. GABA (γ-aminobutyric acid) enrichment and detection methods in cereals: Unlocking sustainable health benefits. Food Chem 2025; 464:141750. [PMID: 39504899 DOI: 10.1016/j.foodchem.2024.141750] [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: 07/25/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
Gamma-aminobutyric acid (GABA), a non-protein amino acid primarily biosynthesized in cereals, is vital to the human neurological system. As the primary neurotransmitter, it promotes relaxation, inhibits depression and sleeplessness, and regulates synaptic transmission and neuronal development. GABA also protects the liver, kidneys, and intestines, reduces blood pressure, and fights cancer and inflammation. Colorimetric and enzyme-based procedures, biosensors, and High-Performance Liquid Chromatography primarily detect GABA. GABA content is enriched by treatments, like germination and fermentation, suitable and economical methods on industrial scales. Moreover, ultrasonication, abiotic stress, heat, and relative humidity are also utilized for GABA enrichment. With advancing technology and knowledge of the nutrients concerning human health, target-specific nutrition utilizing sustainable ingredients needs exploration. Conclusively, the current article delves into the latest developments in GABA enrichment and detection techniques and GABA's potential health benefits. Further studies are required to understand and refine enrichment procedures to develop GABA-enriched foods with health-promoting qualities.
Collapse
Affiliation(s)
- Tabussam Tufail
- School of Food and Biological Engineering Jiangsu University, Zhenjiang, Jiangsu 212013, China; University Institute of Diet and Nutritional Sciences, The University of Lahore, Pakistan
| | - Huma Bader Ul Ain
- University Institute of Diet and Nutritional Sciences, The University of Lahore, Pakistan
| | - Muhammad Safiullah Virk
- School of Food and Biological Engineering Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jawad Ashraf
- School of Food and Biological Engineering Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zahoor Ahmed
- School of Food and Biological Engineering Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, The University of Lahore, Pakistan
| | - Amara Rasheed
- Department of Food Sciences, Government College University Faisalabad, Pakistan
| | - Bin Xu
- School of Food and Biological Engineering Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
6
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2025; 77:1-20. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [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: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Desai SA, Patel VP, Bhosle KP, Nagare SD, Thombare KC. The tumor microenvironment: shaping cancer progression and treatment response. J Chemother 2025; 37:15-44. [PMID: 38179655 DOI: 10.1080/1120009x.2023.2300224] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and treatment response. It comprises a complex network of stromal cells, immune cells, extracellular matrix, and blood vessels, all of which interact with cancer cells and influence tumor behaviour. This review article provides an in-depth examination of the TME, focusing on stromal cells, blood vessels, signaling molecules, and ECM, along with commonly available therapeutic compounds that target these components. Moreover, we explore the TME as a novel strategy for discovering new anti-tumor drugs. The dynamic and adaptive nature of the TME offers opportunities for targeting specific cellular interactions and signaling pathways. We discuss emerging approaches, such as combination therapies that simultaneously target cancer cells and modulate the TME. Finally, we address the challenges and future prospects in targeting the TME. Overcoming drug resistance, improving drug delivery, and identifying new therapeutic targets within the TME are among the challenges discussed. We also highlight the potential of personalized medicine and the integration of emerging technologies, such as immunotherapy and nanotechnology, in TME-targeted therapies. This comprehensive review provides insights into the TME and its therapeutic implications. Understanding the TME's complexity and targeting its components offer promising avenues for the development of novel anti-tumor therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kunal P Bhosle
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Sandip D Nagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kirti C Thombare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| |
Collapse
|
8
|
D'Alessio-Sands L, Gaynier J, Michel-Milian V, Agbowuro AA, Brackett CM. Current Strategies and Future Dimensions in the Development of KRAS Inhibitors for Targeted Anticancer Therapy. Drug Dev Res 2025; 86:e70042. [PMID: 39799558 DOI: 10.1002/ddr.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 12/15/2024] [Indexed: 01/15/2025]
Abstract
KRAS is a proto-oncogene that is found to be mutated in 15% of all metastatic cancers with high prevalence in pancreatic, lung, and colorectal cancers. Additionally, patients harboring KRAS mutations respond poorly to standard cancer therapy. As a result, KRAS is seen as an attractive target for targeted anticancer therapy. Over the last decade, this protein has evolved from being termed "undruggable" to producing two clinically approved drugs along with several more in clinical development, and many under preclinical investigations. This review details the development of various KRAS-targeted molecules with emphasis on the different drug design strategies employed by examining the following areas: (1) Direct inhibition of KRAS mutants using small molecule binders, (2) Inhibiting the activated state of KRAS mutants using a binary complex of small molecule binders and cyclophilin A, and (3) Targeted degradation of KRAS mutants using the PROTAC approach. We assess the pharmacological attributes and possible clinical benefits of the different molecules and look to the next frontiers in the application of KRAS inhibitors as anticancer agents.
Collapse
Affiliation(s)
| | - Joshua Gaynier
- South University School of Pharmacy, Savannah, Giorgia, USA
| | | | | | | |
Collapse
|
9
|
Eixenberger JE, Anders CB, Hermann R, Wada K, Reddy KM, Montenegro-Brown RJ, Fologea D, Wingett DG. On-demand release of encapsulated ZnO nanoparticles and chemotherapeutics for drug delivery applications. RSC PHARMACEUTICS 2025; 2:82-93. [PMID: 39703205 PMCID: PMC11650639 DOI: 10.1039/d4pm00189c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/17/2024] [Indexed: 12/21/2024]
Abstract
Nanomedicines offer high promise for the treatment of various diseases, and numerous novel approaches using nanomaterials have been developed over the years. In this report, we introduce a new strategy utilizing ZnO nanoparticles (nZnO) to trigger the rapid release of lipid-encapsulated therapeutics upon photo-irradiation with UV light (365 nm). In vitro studies demonstrate that encapsulation of nZnO effectively eliminates the cytotoxicity of nZnO, but this can be re-established upon release from the lipid coating. Using 5(6)-carboxyfluorescein as a model for hydrophilic drug loading, we show the ability to co-load drugs with nZnO into liposomes. Kinetic studies reveal the ability to release the majority of the dye within 60 minutes post-photo-irradiation and provide insights into factors that impact release kinetics. To further explore this, Jurkat T cell leukemia and T47D breast cancer cells were treated with co-encapsulated nZnO and the hydrophobic cancer drug paclitaxel. These studies revealed enhanced toxicity of the triggered release groups with an extreme difference noted in the viability profiles of the T47D breast cancer cell model. Taken together, these studies indicate that this system of co-encapsulating nZnO and chemotherapeutic drugs has the potential to minimize systemic toxicity, by controlling therapeutic release, while allowing for the localized selective destruction of cancer.
Collapse
Affiliation(s)
- Josh E Eixenberger
- Biomolecular Sciences Graduate Program, Boise State University Boise ID 83725 USA +208-426-2231
- Department of Physics, Boise State University Boise ID 83725 USA
| | - Catherine B Anders
- Biomolecular Sciences Graduate Program, Boise State University Boise ID 83725 USA +208-426-2231
| | - Rebecca Hermann
- Department of Biological Sciences, Boise State, University Boise ID 83725 USA +208-426-2921
| | - Katelyn Wada
- Department of Physics, Boise State University Boise ID 83725 USA
| | - Kongara M Reddy
- Department of Physics, Boise State University Boise ID 83725 USA
| | | | - Daniel Fologea
- Biomolecular Sciences Graduate Program, Boise State University Boise ID 83725 USA +208-426-2231
- Department of Physics, Boise State University Boise ID 83725 USA
| | - Denise G Wingett
- Biomolecular Sciences Graduate Program, Boise State University Boise ID 83725 USA +208-426-2231
- Department of Biological Sciences, Boise State, University Boise ID 83725 USA +208-426-2921
| |
Collapse
|
10
|
Sharma U, Sahni PK, Sharma B, Gupta M, Kaur D, Mathkor DM, Haque S, Khatoon S, Tuli HS, Mishra A, Ahmad F. Silymarin: a promising modulator of apoptosis and survival signaling in cancer. Discov Oncol 2025; 16:66. [PMID: 39836338 PMCID: PMC11751200 DOI: 10.1007/s12672-025-01800-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Cancer, one of the deadliest diseases, has remained the epicenter of biological research for more than seven decades. Yet all the efforts for a perfect therapeutic cure come with certain limitations. The use of medicinal plants and their phytochemicals as therapeutics has received much attention in recent years. Silymarin, a polyphenolic flavonoid with a variety of anti-cancerous properties, was isolated from the plant Silybum marianum. The present review centres on the function of silymarin in controlling important signalling pathways related to apoptosis and survival, such as the JAK/STAT pathway, PI3K/Akt/mTOR, Bcl-2/Bax, and Fas/FasL. It is emphasised that silymarin's capacity to target these pathways is a key mechanism behind its anticancer effects against a variety of malignancies. By upregulating pro-apoptotic and downregulating anti-apoptotic proteins, silymarin controls a series of events that result in tumor suppression and cell death in a variety of cancer types. The low bioavailability and limited therapeutic efficacy of silymarin are improved by the application of various nano-delivery systems. As efficient carriers, liposomes, polymeric micelles, lipid- and metal-based nanoparticles, increase the solubility and distribution of silymarin in target tissues. Lastly, a number of preclinical studies that provide a basis for upcoming therapeutic interventions are highlighted in the review, providing encouraging directions for additional research and advancement.
Collapse
Affiliation(s)
- Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Praveen Kumar Sahni
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Bunty Sharma
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, Uttarakhand, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Damandeep Kaur
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, 140413, India
| | - Darin Mansor Mathkor
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
| | - Shafiul Haque
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
- Universidad Espiritu Santo, Samborondon, Ecuador
| | - Sabiha Khatoon
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Astha Mishra
- Department of Optometry, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, India
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, India.
| |
Collapse
|
11
|
Atefeh M, Elizabeth M, Patricia N. Medication Management of Patients With Cancer Undergoing Surgery From Preadmission to Discharge: A Mixed-Methods Systematic Review. J Adv Nurs 2025. [PMID: 39835655 DOI: 10.1111/jan.16759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
AIM(S) To identify and synthesise available evidence about regular medication management processes, from preadmission to discharge from hospital, in patients with cancer undergoing surgery. DESIGN Mixed-methods systematic review. METHODS Studies published from inception of each database until February 2023 were screened, utilising four main search concepts. The JBI methodology for mixed-methods systematic review was followed in this review. DATA SOURCES MEDLINE, CINAHL, EMBASE, APA PsycINFO, Scopus and Web of Science. RESULTS Eight out of 717 screened studies were included. Two themes related to patients' medication management were identified. Preoperative factors such as polypharmacy, potentially inappropriate medications, delirium-inducing medications and preoperative discontinuation-requiring medications were associated with several postoperative complications in patients with cancer. Additionally, pharmacist-led interventions and collaborative efforts between nurses and patients were shown to improve the medication management process across the perioperative pathway. CONCLUSION This systematic review emphasises the necessity of effectively managing regular medication, especially before surgery, to mitigate postoperative complications in patients with cancer. It offers critical insights into how involving pharmacists and nurses enhances medication management outcomes, benefiting health care professionals and institutions aiming to optimise perioperative medication therapy. IMPLICATIONS FOR THE PROFESSION AND/OR PATIENT CARE Enhancing patients' regular medication management through comprehensive reviews before surgery, and improving collaborative practices among pharmacists, nurses and patients via targeted interventions introduced by health care organisations, ensure safe medication use throughout the perioperative pathway. IMPACT Improving regular medication management process can reduce risk of medication errors and adverse drug events and enhance postoperative outcomes. REPORTING METHOD SWiM reporting guidelines. PATIENT OR PUBLIC CONTRIBUTION No patient or public contribution.
Collapse
Affiliation(s)
- Mehrabifar Atefeh
- School of Nursing and Midwifery, Centre for Quality and Patient Safety Research, Institute for Health Transformation, Deakin University, Geelong, Victoria, Australia
| | - Manias Elizabeth
- School of Nursing and Midwifery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Nicholson Patricia
- School of Nursing and Midwifery, Centre for Quality and Patient Safety Research, Institute for Health Transformation, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
12
|
Jain A, Sivakumar N, Sharma P, John S, Gupta S. TNF-α polymorphism in oral cancer and oral potentially malignant disorders: meta-analysis and investigation as a potential tool to determine individual susceptibility and the prognosis. Int J Oral Maxillofac Surg 2025:S0901-5027(25)00002-5. [PMID: 39827024 DOI: 10.1016/j.ijom.2025.01.002] [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: 03/30/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
There is increasing evidence that tumour necrosis factor alpha (TNF-α) gene polymorphisms may promote development and spread of cancer through transitions at different loci that lead to its altered expression. However, data on TNF-α gene polymorphisms in oral cancer remain highly controversial. This meta-analysis was performed to determine the impact of TNF-α gene polymorphisms on oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMD). An electronic search was conducted in the PubMed, Cochrane Library, Google Scholar, and Web of Science databases to identify original articles reporting TNF-α polymorphism in OSCC and OPMD. The articles were examined by two independent authors. Data from the included articles were compiled and tabulated. Risk of bias was analysed. Overall, 11 eligible articles that included 1070 cases of OSCC and 641 cases of OPMD in which TNF-α polymorphism was studied, were included for quantitative analysis. It was found that both TNF-α -308 and -238 polymorphisms were significantly associated with the development of oral cancer. TNF-α polymorphism was significantly associated with OSCC and OPMD. Moreover, in polymorphisms of TNF-α at both -308 and -238, the G allele, especially the homozygous form (GG), was found to be associated with oral cancer.
Collapse
Affiliation(s)
- A Jain
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow, UP, India.
| | - N Sivakumar
- Department of Oral Pathology and Microbiology, CDER, AIIMS, New Delhi, India.
| | - P Sharma
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow, UP, India.
| | - S John
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow, UP, India.
| | - S Gupta
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow, UP, India.
| |
Collapse
|
13
|
Mosallam G, Winer ES, Keating JH, Flamand Y, Solodokin LJ. Utility of ursodiol prophylaxis against sinusoidal obstruction syndrome (SOS)/ veno-occlusive disease (VOD) in acute leukemia patients receiving gemtuzumab-ozogamicin (GO) or inotuzumab-ozogamicin (InO). J Oncol Pharm Pract 2025:10781552241313473. [PMID: 39819278 DOI: 10.1177/10781552241313473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
PURPOSE Sinusoidal obstructive syndrome (SOS)/veno-occlusive disease (VOD) is a serious complication in hematopoietic stem-cell transplant (HSCT) patients. Gemtuzumab-ozogamicin (GO) and InO are known to cause SOS/VOD in leukemic and transplant populations. Due to limited data on ursodiol prophylaxis in non-HSCT patients, we aimed to assess hepatotoxicity, SOS/VOD incidences, time to hepatotoxicity, and confirmed SOS/VOD in adults receiving GO or InO ± ursodiol. METHODS A multicenter, retrospective chart review of adult acute leukemia patients who received ≥1 dose of GO or InO at DFCI/some of the Harvard Cancer Centers during 4-year period (9/1/2017-9/1/2021). Acute promyelocytic leukemia patients and post-GO or InO HSCT-recipients (100-day follow-up period) were excluded. Descriptive summaries are provided, direct comparisons were made using Student T-test (continuous variables) and Fisher's exact test (categorical variables). RESULTS In our population (N = 82), 87.8% received ursodiol and 12.2% did not. There were no significant differences in baseline to peak hepatic labs. The No-Ursodiol Group had higher incidence of Grade 3 aspartate aminotransferase (AST) transaminitis vs. the Ursodiol Group (60% vs. 20.8%; p = 0.015), and a trend towards shorter mean time to Grade 3 AST transaminitis (18.5 vs. 23.8 days; p = 0.30). Moreover, 4.2% of Ursodiol Group developed SOS/VOD vs. 0% in the No-Ursodiol Group (NS). Three patients developed SOS/VOD: 2 received GO, 1 received InO, and 2 were alive by the end of the follow-up period. CONCLUSION In our cohort, ursodiol prophylaxis in adults receiving GO/InO is not associated with lower incidences of hepatotoxicity, SOS/VOD, or time to Grade 3 AST transaminitis, but is associated with decreased incidence of AST elevations.
Collapse
Affiliation(s)
- Grace Mosallam
- Department of Pharmacy Practice, Massachusetts College of Pharmacy and Health Sciences School of Pharmacy, Boston, MA, USA
| | - Eric S Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Julia H Keating
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yael Flamand
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Loriel J Solodokin
- Department of Pharmacy Practice, Massachusetts College of Pharmacy and Health Sciences School of Pharmacy, Boston, MA, USA
- Department of Pharmacy Services, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
14
|
Kangra K, Kakkar S, Mittal V, Kumar V, Aggarwal N, Chopra H, Malik T, Garg V. Incredible use of plant-derived bioactives as anticancer agents. RSC Adv 2025; 15:1721-1746. [PMID: 39835210 PMCID: PMC11744461 DOI: 10.1039/d4ra05089d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Cancer is a major global concern. Despite considerable advancements in cancer therapy and control, there are still large gaps and requirements for development. In recent years, various naturally occurring anticancer drugs have been derived from natural resources, such as alkaloids, glycosides, terpenes, terpenoids, flavones, and polyphenols. Plant-derived substances exhibit their anticancer potential through antiproliferative activity, cytotoxicity, apoptosis, angiogenesis and cell cycle arrest. Natural compounds can affect the molecular activity of cells through various signaling pathways, like the cell cycle pathway, STAT-3 pathway, PI3K/Akt, and Ras/MAP-kinase pathways. Capsaicin, ouabain, and lycopene show their anticancer potential through the STAT-3 pathway in breast, colorectal, pancreatic, lung, cervical, ovarian and colon cancers. Epigallocatechin gallate and emodin target the JNK protein in skin, breast, and lung cancers, while berberine, evodiamine, lycorine, and astragalin exhibit anticancer activity against breast, liver, prostate, pancreatic and skin cancers and leukemia through the PI3K/Akt and Ras/MAP-kinase pathways. In vitro/in vivo investigations revealed that secondary metabolites suppress cancer cells by causing DNA damage and activating apoptosis-inducing enzymes. After a meticulous literature review, the anti-cancer potential, mode of action, and clinical trials of 144 bioactive compounds and their synthetic analogues are included in the present work, which could pave the way for using plant-derived bioactives as anticancer agents.
Collapse
Affiliation(s)
- Kiran Kangra
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Saloni Kakkar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| | - Virender Kumar
- College of Pharmacy, Pandit Bhagwat Dayal Sharma University of Health Sciences Rohtak 124001 India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University) Mullana Ambala 133207 Haryana India
| | - Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences Chennai 602105 Tamil Nadu India
| | - Tabarak Malik
- Department of Biomedical Sciences, Jimma University Jimma Ethiopia
- Division of Research & Development, Lovely Professional University Phagwara Punjab-144411 India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University Rohtak 124001 India
| |
Collapse
|
15
|
Bhat AM, Bhat IA, Malik MA, Kaiser P, Ramajayan P, Rayees SR, Ahmed Z, Tasduq SA. Inhibition of IKK complex by (2 methyl butyryl) Shikonin, a naturally occurring naphthoquinone, abrogates melanoma growth and progression via modulation of the IKK/NFκB /EMT signaling axis. Int Immunopharmacol 2025; 148:114026. [PMID: 39823792 DOI: 10.1016/j.intimp.2025.114026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
Melanoma is an aggressive form of malignancy that originates from melanin-producing cells known as melanocytes underlying the basal layer of the epidermis with a poor prognosis, low survival rates, and limited treatment options. Although several specific and effective systematic strategies for treating melanoma have been established, the underlying molecular mechanism of melanoma progression, mortality and the promising therapeutic options remain elusive. Shikonin (SK), a natural naphthoquinone derived from a medicinal herbaceous plant, has been shown to inhibit the proliferation of several cancer cells. However, its role in the context of melanoma is poorly understood. In the present study, the anti-melanoma activity of (2-methylbutyryl) Shikonin was assessed under in vitro and in vivo models. In vitro findings revealed that (2-methylbutyryl) Shikonin significantly reduced the viability and promoted apoptosis in the B16F10 melanoma cells. Additionally (2-methylbutyryl) Shikonin significantly suppressed migration and invasion of melanoma cells by regulating IKK/NFκB/EMT signalling axis thereby attenuating nuclear translocation and subsequent transcription of NF-κB downstream target genes. Furthermore, (2-methylbutyryl) Shikonin administration significantly reduced tumor size and weight in the xenograft melanoma mice model. Our data presents novel insights that justify additional preclinical and clinical validations of (2-methylbutyryl) Shikonin for melanoma therapy.
Collapse
Affiliation(s)
- Aalim Maqsood Bhat
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Irshad Ahmad Bhat
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Mushtaq Ahmad Malik
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Peerzada Kaiser
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - P Ramajayan
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sheikh R Rayees
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Zabeer Ahmed
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sheikh Abdullah Tasduq
- Pharmacology Division, Council of Scientific and Industrial Research-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
16
|
Aqib M, Khatoon S, Ali M, Sajid S, Assiri MA, Ahamad S, Saquib M, Hussain MK. Exploring the anticancer potential and mechanisms of action of natural coumarins and isocoumarins. Eur J Med Chem 2025; 282:117088. [PMID: 39608206 DOI: 10.1016/j.ejmech.2024.117088] [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: 10/21/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
Natural coumarins and isocoumarins show significant therapeutic potential against cancer in preclinical studies by targeting multiple pathways and processes. These compounds influence several critical cellular processes, such as apoptosis, autophagy, and cell cycle regulation, which are pivotal in cancer development and progression. Their capability to target multiple signalling pathways provides a strategic advantage over single-target therapies, which are often limited by drug resistance. Notably, coumarins have the potential to inhibit angiogenesis, the process through which tumours develop new blood vessels, thereby potentially restricting tumour growth and metastasis. Additionally, coumarins may enhance anticancer effects by modulating immune responses and reducing inflammation, thus offering a dual approach to combating cancer. They also show promise in addressing multidrug resistance, a significant challenge in cancer therapy, by targeting drug efflux proteins and potentially improving the efficacy of existing treatments. While preclinical studies are promising, further research is required to elucidate the pharmacokinetics, toxicity, and potential side effects of coumarins in humans. Continued clinical evaluation will be crucial to confirm their effectiveness in cancer patients. Nonetheless, their ability to target multiple pathways positions coumarin based molecules as potential candidates for future anti-cancer drug development.
Collapse
Affiliation(s)
- Mohd Aqib
- Department of Chemistry, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | | | - Mujahid Ali
- Department of Physical Education, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | - Shabana Sajid
- Department of Chemistry, Gandhi Faiz-e-Aam College, Shahjahanpur, M. J. P. Rohilkhand University, Bareilly, UP, India
| | - Mohammed Ali Assiri
- Research Center for Advanced Materials Science (RCAMS), Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha, 61413, Saudi Arabia
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad), 211002, UP, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad), 211010, UP, India.
| | - Mohd Kamil Hussain
- Department of Chemistry, Govt. Raza P.G. College, Rampur, M. J. P. Rohilkhand University, Bareilly, UP, India.
| |
Collapse
|
17
|
Aparna TN, Kumar R, Ali SR, Patel DJ, Julekha K, Begum T, Bala J, Kumar P. Silica Nanoparticles: A Promising Vehicle for Anti-Cancer Drugs Delivery. AAPS PharmSciTech 2025; 26:33. [PMID: 39806209 DOI: 10.1208/s12249-024-02982-9] [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: 08/27/2024] [Accepted: 10/24/2024] [Indexed: 01/16/2025] Open
Abstract
The prevalence and death due to cancer have been rising over the past few decades, and eliminating tumour cells without sacrificing healthy cells remains a difficult task. Due to the low specificity and solubility of drug molecules, patients often require high dosages to achieve the desired therapeutic effects. Silica nanoparticles (SiNPs) can effectively deliver therapeutic agents to targeted sites in the body, addressing these challenges. Using SiNPs as vehicles for anti-cancer drug delivery has emerged as a promising strategy due to their unique structural properties, biocompatibility, and versatility. This review explores the various aspects of SiNPs in cancer therapy, highlighting their synthesis, functionalization, and application in delivering chemotherapeutic agents, photosensitizers, and nucleic acids. SiNPs offer advantages such as high drug loading capacity, controlled release, and targeted delivery, enhancing therapeutic efficacy and reducing systemic toxicity. Moreover, this review aims to provide an in-depth understanding of the current state and prospects of SiNPs in revolutionizing cancer treatment and improving patient outcomes.
Collapse
Affiliation(s)
- T Naga Aparna
- Department of Pharmaceutics, G. Pullareddy College of Pharmacy, Mehdipatnam, Hyderabad, India
| | - Rohit Kumar
- Department of Pharmaceutics, RKSD College of Pharmacy, Ambala Road, Kaithal, 136027, Haryana, India
| | - Shah Raj Ali
- Department of Chemistry, D.S.B. Campus, Kumaun University, Nainital, 263002, Uttarakhand, India
| | - Dhaval J Patel
- Department of Pharmaceutics, Saraswati Institute of Pharmaceutical Sciences, Dhanap, Gandhinagar, 382354, Gujarat, India
| | - Kazi Julekha
- Department of Pharmaceutical Technology, Brainware University, Barasat, Kolkata, 700125, West Bengal, India
| | - Touseef Begum
- Department of Pharmaceutical Sciences, Ibn Sina National College for Medical Studies, Jeddah, 31906, Kingdom of Saudi Arabia
| | - Jyoti Bala
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India
| | - Pawan Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India.
| |
Collapse
|
18
|
Malek S, Jaafari MR, Mahmoudi A, Mohammadi M, Malaekeh-Nikouei B. Smart release injectable hydrogel co-loaded with liposomal combretastatin A4 and doxorubicin nanogel for local combinational drug delivery: A preclinical study. Int J Pharm 2025; 671:125213. [PMID: 39814242 DOI: 10.1016/j.ijpharm.2025.125213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/21/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Surgical resection and postoperative adjuvant chemotherapy have enhanced the outlook for breast cancer patients. However, tumor relapse and serious side effects of chemotherapy continue to impact patients' quality of life. Designing injectable composite hydrogel made of biodegradable polymers providing sustained release of antiangiogenic and chemotherapeutic agents might play a vital role in elimination of cancer cells. In this regard, we developed dextran based composite hydrogel incorporating doxorubicin-loaded dual-sensitive pH-redox nanogels (DOX-DSNG) and combretastatin A4 (CA4) loaded liposomes which undergo rapid disassembly in cancer cells. CA4 prevents tubulin polymerization and thus inhibits angiogenesis by binding to vascular endothelial tubulin. The results showed that DOX-DSNGs were negatively charged and 144.8 ± 0.85 nm in size. Besides, the size of CA4 loaded liposomes were 102.35 ± 4.22 nm and were negatively charged. Encapsulation efficiency of DOX-DSNGs and CA4 loaded liposomes were 100 % and 89 %, respectively. After loading into the hydrogel structure, doxorubicin and CA4 were gradually released from the composite hydrogel for up to 21 days. DOX-DSNGs and CA4 loaded liposomes showed a dose-dependent cytotoxic effect against 4 T1 breast cancer cells. Thereafter, the anti-neoplastic effect and survival study of the composite hydrogel was evaluated in vivo in tumor-bearing mice. The composite hydrogel significantly reduced tumor volume (from 116 mm3 to 38 mm3) with negligible organ damage, while showed lower cardiotoxicity in 28 days. In conclusion, our results revealed that injectable composite dextran-based hydrogel incorporated with DOX-DSNG and CA4 loaded liposomes could be used as an efficient delivery platform for combination therapies in treatment of solid tumors.
Collapse
Affiliation(s)
- Saba Malek
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mahmoudi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Bizhan Malaekeh-Nikouei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Rodríguez-Machuca VU, Ángulo-De La Cruz EJ, Gaxiola-Sarmiento AL, Ramírez-Corona JA, Fernández-Sánchez D, Arredondo-Navarro LA, Velázquez-Santana H, Nuñez-Velasco S, Navarro-Martín Del Campo RM, Bustos-Rodríguez FDJ, Corona-Rivera JR, Bobadilla-Morales L, Corona-Rivera A. Long-term epidemiological trends in (primary) pediatric central nervous system tumors: a 25-year cohort analysis in Western Mexico. Childs Nerv Syst 2025; 41:90. [PMID: 39798037 DOI: 10.1007/s00381-024-06715-y] [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: 08/29/2024] [Accepted: 12/06/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND Central nervous system tumors (CNSTs) represent a significant oncological challenge in pediatric populations, particularly in developing regions where access to diagnostic and therapeutic resources is limited. METHODS This research investigates the epidemiology, histological classifications, and survival outcomes of CNST in a cohort of pediatric patients aged 0 to 19 years within a 25-year retrospective study at the Civil Hospital of Guadalajara, Mexico, from 1999 to 2024. RESULTS Data was analyzed from 273 patients who met inclusion criteria, revealing a higher incidence in males (51.6%) with a mean age at diagnosis of 8.2 years. Histological analysis revealed gliomas as the most common type (52.7%), followed by embryonal tumors (28.6%). High-grade tumors (WHO grade 4) comprised 49.8% of cases, demonstrating significantly poorer survival outcomes (median overall survival of 13.5 months) compared to lower-grade tumors (up to 57 months). The predominance of tumors in the supratentorial region and the notable differences in survival outcomes by tumor type underscore the varied impact of geographical and socioeconomic factors on pediatric oncology in Mexico. CONCLUSION This study highlights the critical need for improved healthcare infrastructure and early diagnosis initiatives, as well as the importance of targeted research to address disparities in treatment and outcomes for pediatric CNST in this region.
Collapse
Affiliation(s)
- Víctor Ulises Rodríguez-Machuca
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Cytogenetics Unit, Treatment Diagnostic and Auxiliary Division, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | | | | | - Juan Antonio Ramírez-Corona
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Cytogenetics Unit, Treatment Diagnostic and Auxiliary Division, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | - David Fernández-Sánchez
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Cytogenetics Unit, Treatment Diagnostic and Auxiliary Division, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | | | | | | | | | - Felipe De Jesús Bustos-Rodríguez
- University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Pathology Service, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | - Jorge Román Corona-Rivera
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Pediatrics Division, Center for Registry and Research in Congenital Anomalies (CRIAC), Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | - Lucina Bobadilla-Morales
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
- Cytogenetics Unit, Treatment Diagnostic and Auxiliary Division, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico
| | - Alfredo Corona-Rivera
- Ph.D. Human Genetics Program, Molecular Biology and Genomics Department, Human Genetics Institute "Dr. Enrique Corona-Rivera", University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico.
- Cytogenetics Unit, Treatment Diagnostic and Auxiliary Division, Civil Hospital "Dr. Juan I. Menchaca", Guadalajara, Mexico.
| |
Collapse
|
20
|
Sun D, Okosun BO, Xue Y, Tayutivutikul K, Smith KH, Darland DC, Zhao JX. Multi-functional near-infrared fluorescent polymer dot-siRNA for gene expression regulation. J Mater Chem B 2025. [PMID: 39784355 DOI: 10.1039/d4tb01954g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Regulation of gene expression in eukaryotic cells is critical for cell survival, proliferation, and cell fate determination. Misregulation of gene expression can have substantial, negative consequences that result in disease or tissue dysfunction that can be targeted for therapeutic intervention. Several strategies to inhibit gene expression at the level of mRNA transcription and translation have been developed, such as anti-sense inhibition and CRISPR-Cas9 gene editing. However, these strategies have some limitations in terms of specificity, toxicity, and ease of use. We have designed a nanomaterials-based tool to inhibit gene expression in eukaryotic cells with a potential application in basic and biomedical research. At the heart of our rational design approach is a polymer dots (Pdots)-based nanoplatform that can provide a means to deliver gene-specific small interfering (siRNA) into cells while at the same time providing a visualization mechanism to determine which cells have taken up the siRNA. The Pdots that we designed and synthesized had an average size 64.25 ± 0.60 nm and a zeta potential that was +37.40 ± 8.28 mV. The Pdot-1 nmole Gapdh siRNA showed an average size of 82.27 ± 9.83 nm, with the zeta potential values determined to be -52.00 ± 6.05 mV in the HEPES buffer. Both Pdots and Pdot-siRNA displayed two emission peaks in the visible (588 nm) and near-infrared (NIR) emission range (775 nm). We treated primary cultures of mouse brain-derived microvascular cells with Pdot-Gapdh siRNA and observed uniform cellular uptake of the nanomaterial in the cells and reduced intensity of Gapdh immunolabeling. Our results highlight the potential application of Pdot-siRNA for gene expression targeting with simultaneous visual monitoring of Pdot-siRNA delivery. The simple design offers a flexible and novel strategy to inhibit a wide range of mRNA targets with minimal toxicity, high efficiency, and focused cell visualization.
Collapse
Affiliation(s)
- Di Sun
- Department of Chemistry, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Blessing O Okosun
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Yujie Xue
- Department of Chemistry, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Kirati Tayutivutikul
- Department of Chemistry, University of North Dakota, Grand Forks, ND, 58202, USA.
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Kaitlyn H Smith
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Julia X Zhao
- Department of Chemistry, University of North Dakota, Grand Forks, ND, 58202, USA.
| |
Collapse
|
21
|
Moinuddin SK, Srikanth P, Sharma P, Nandi S. Bioanalytical Method Development and Validation of Doxorubicin and Lapatinib in Rat Plasma Using UHPLC-HESI-LTQ-MS. J Chromatogr Sci 2025; 63:bmad090. [PMID: 38088022 DOI: 10.1093/chromsci/bmad090] [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: 09/16/2023] [Revised: 11/15/2023] [Indexed: 01/11/2025]
Abstract
Cancer is considered a silent killer. The complexity of cancer makes it earn that title. So far there are only a few approaches to treat cancer. Among them, chemotherapy is considered the best approach. Many chemotherapeutical compounds are commercially available in the market. Among them, doxorubicin (DOX) and lapatinib (LAP) are considered blockbuster molecules. However, DOX suffers from poor bioavailability and exhibits cardiotoxicity. Interestingly, a fixed dose combination of DOX and LAP significantly decreases the cardiotoxic effect of DOX. To enhance the oral bioavailability of DOX and to avail the synergistic effect of LAP, many formulations have been made. To quantify both compounds in any formulation or biological matrix, an Liquid chromatography-Mass Spectrometry (LC-MS) method is required. In this present study, a simple and rapid Ultra High-Performance Liquid Chromatography - Heated Electron Spray Ionization - Mass Spectrometry (UHPLC-HESI-MS) bioanalytical method was developed. The developed method was validated as per the regulatory guidelines. The validated bioanalytical method had a lower limit of quantification of 0.75 ng. A simple protein precipitation technique was optimized to extract the compounds from the rat plasma. All the validation parameters were found to be within the limits as per the regulatory guidelines. A novel and rapid analytical method was successfully developed and validated. This developed method can be used to quantify the DOX and LAP in any formulation and biological matrix.
Collapse
Affiliation(s)
- Shaik Khaja Moinuddin
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Pirangi Srikanth
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Parul Sharma
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Sukhendu Nandi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160 062, India
| |
Collapse
|
22
|
Wang H, Ma X, Sun L, Bi T, Yang W. Applications of innovative synthetic strategies in anticancer drug discovery: The driving force of new chemical reactions. Bioorg Med Chem Lett 2025; 119:130096. [PMID: 39798856 DOI: 10.1016/j.bmcl.2025.130096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
The discovery of novel anticancer agents remains a critical goal in medicinal chemistry, with innovative synthetic methodologies playing a pivotal role in advancing this field. Recent breakthroughs in CH activation reactions, cyclization reactions, multicomponent reactions, cross-coupling reactions, and photo- and electro-catalytic reactions have enabled the efficient synthesis of new molecular scaffolds exhibiting potent biological activities, including anticancer properties. These methodologies have facilitated the functionalization of natural products, the modification of bioactive molecules, and the generation of entirely new compounds, many of which demonstrate strong antitumor activity. This review summarizes the latest synthetic strategies employed over the past five years for discovering anticancer agents, focusing on their influence on drug design. Additionally, the role of new chemical reactions in expanding chemical space and overcoming challenges, such as drug resistance and selectivity, is highlighted, further emphasizing the importance of discovering novel reactions as a key trend in future drug development.
Collapse
Affiliation(s)
- Han Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolong Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Longkang Sun
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Tongyu Bi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weibo Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
23
|
Bhandoria GP, Jayraj AS, Tiwari S, Migliorelli F, Nelson G, van Ramshorst GH, Kacperczyk-Bartnik J, Angeles MA, Nair N, El Hajj H, Bizzarri N. Use of social media for academic and professional purposes by gynecologic oncologists. Int J Gynecol Cancer 2025:ijgc-2024-005573. [PMID: 38955373 DOI: 10.1136/ijgc-2024-005573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE To investigate the prevalence and patterns of social media use among gynecologic oncologists for professional and academic purposes. METHODS A prospective online survey between November and December 2022 targeted gynecologic oncology practitioners (gynecologic oncologists, surgical oncologists, medical oncologists, radiation/clinical oncologists, and onco-pathologists/pathologists). The survey, distributed via various social media platforms, included 40 questions to capture qualitative and quantitative data on social media use. RESULTS Of 131 respondents from 32 countries, 106 (80.9%) were gynecologic oncologists and affiliated with academic institutions (84.7%). Facebook (n=110, 83.9%), Twitter (n= 108, 82.4%), and Instagram (n=100, 76.3%) were the most used platforms. Respondents used social media to stay updated (n=101, 77.1%), network (n=97, 74%), learn about conferences and webinars (n=97, 74%), and engage in academic discussions (n=84, 64.1%). Following the COVID-19 pandemic, 100/129 (77.5%) reported increased social media use. However, only 32 (24.4%) used it to connect with patients, and concerns were raised about privacy and the need for separate professional and personal accounts. A quarter of respondents hesitated to share their opinions on social media due to the fear of controversy, with 26 (20%) experiencing cyberbullying, yet 120/130 (92.3%) believed it enabled junior professionals to express their views. Concerns about differentiating valid content, information reliability, and the professional perception of sourcing knowledge from social media were noted. Gender, age, specialty, and income level influenced patterns of social media use, with variations in preferences for platforms, content engagement, and purposes, highlighting a complex landscape of social media interaction among gynecologic oncologists. CONCLUSION While the use of social media among gynecologic oncologists is prevalent, particularly for academic and professional development, challenges such as cyberbullying, privacy concerns, and the need for formal training in social media navigation persist. Tailored training programs and guidelines could enhance social media's effective and ethical use in this field, promoting a safe environment for professional expression and engagement.
Collapse
Affiliation(s)
- Geetu Prakash Bhandoria
- Department of Obstetrics and Gynecology, Command Hospital Kolkata, Kolkata, West Bengal, India
| | - Aarthi S Jayraj
- South Tees NHS Foundation Trust, James Cook University, Middlesbrough, UK
| | - Shyamji Tiwari
- Department of Obstetrics and Gynaecology, Military Hospital Patiala, Patiala, Punjab, India
| | - Federico Migliorelli
- Maternal-Fetal Medicine Department, Hospital Clinic Barcelona, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Barcelona, Spain
| | - Gregg Nelson
- Department of Obstetrics and Gynecology, University of Calgary, Calgary, Alberta, Canada
| | - Gabriëlle H van Ramshorst
- Department of Gastrointestinal Surgery, University Hospital Ghent, Ghent, Belgium
- Human Structure and Repair, Ghent University, Ghent, Belgium
| | | | - Martina Aida Angeles
- Department of Gynecologic Oncology, Centro Sociosanitario y Residencial de la Vall d'Hebron de Barcelona, Barcelona, Spain
| | - Navya Nair
- Division of Gynecologic Oncology, Dept of Obstetrics and Gynecology, University of Miami Sylvester Cancer Center, Miami, Florida, USA
| | - Houssein El Hajj
- Department of Gynecologic Oncology, Institute Curie, Paris, France
| | - Nicolò Bizzarri
- UOC Ginecologia Oncologica, Dipartimento per la salute della Donna e del Bambino e della Salute Pubblica, Policlinico Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
24
|
Costa AF, Teixeira A, Reis CA, Gomes C. Novel anticancer drug discovery efforts targeting glycosylation: the emergence of fluorinated monosaccharides analogs. Expert Opin Drug Discov 2025:1-11. [PMID: 39749684 DOI: 10.1080/17460441.2024.2444375] [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: 09/05/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Glycosylation is an essential enzymatic process of building glycan structures that occur mainly within the cell and gives rise to a diversity of cell surface and secreted glycoconjugates. These glycoconjugates play vital roles, for instance in cellcell adhesion, interaction and communication, activation of cell surface receptors, inflammatory response and immune recognition. This controlled and wellcoordinated enzymatic process is altered in cancer, leading to the biosynthesis of cancerassociated glycans, which impact glycandependent biological roles. AREAS COVERED In this review, the authors discuss the importance of targeting cancerassociated glycans through potent glycan biosynthesis inhibitors. It focuses on the use of analogs, providing an overview of findings involving these in cancer. The highly explored fluorinated monosaccharide analogs targeting aberrant glycosylation are described, aiming to inspire advances in the field. EXPERT OPINION Altered glycosylation, such as increased sialylation and fucosylation, is a feature in cancer and has been shown to play key roles in several malignant properties of cancer cells. Strategies aiming at remodeling cancer cells´ glycome are emerging and present a huge potential for cancer therapy. Fluorinated monosaccharides have been gathering promising preclinical results as novel cancer drugs. Nevertheless, cancer specific targeting strategies must be considered to avoid significant sideeffects.
Collapse
Affiliation(s)
- Ana F Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Porto, Portugal
| | - Andreia Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Celso A Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
| | - Catarina Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| |
Collapse
|
25
|
Lu Y, Li Z, Zhu X, Zeng Q, Liu S, Guan W. Novel Modifications and Delivery Modes of Cyclic Dinucleotides for STING Activation in Cancer Treatment. Int J Nanomedicine 2025; 20:181-197. [PMID: 39802380 PMCID: PMC11721825 DOI: 10.2147/ijn.s503780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025] Open
Abstract
The microenvironment tends to be immunosuppressive during tumor growth and proliferation. Immunotherapy has attracted much attention because of its ability to activate tumor-specific immune responses for tumor killing. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is an innate immune pathway that activates antitumor immunity by producing type I interferons. Cyclic dinucleotides (CDNs), produced by cGAS sensing cytoplasmic abnormal DNA, are major intermediate activating molecules in the STING pathway. Nowadays, CDNs and their derivatives have widely worked as powerful STING agonists in tumor immunotherapy. However, their clinical translation is hindered by the negative electrical properties, sensitivity to hydrolytic enzymes, and systemic toxicity. Recently, various CDN delivery systems have made significant progress in addressing these issues, either through monotherapy or in combination with other treatment modalities. This review details recent advances in CDNs-based pharmaceutical development or delivery strategies for enriching CDNs at tumor sites and activating the STING pathway.
Collapse
Affiliation(s)
- Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Zhiyan Li
- Division of Thoracic Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xudong Zhu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Qingwei Zeng
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Song Liu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Wenxian Guan
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| |
Collapse
|
26
|
Vigna V, Cova TFGG, Pais AACC, Sicilia E. Prediction of Pt, Ir, Ru, and Rh complexes light absorption in the therapeutic window for phototherapy using machine learning. J Cheminform 2025; 17:1. [PMID: 39757232 DOI: 10.1186/s13321-024-00939-5] [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: 08/22/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025] Open
Abstract
Effective light-based cancer treatments, such as photodynamic therapy (PDT) and photoactivated chemotherapy (PACT), rely on compounds that are activated by light efficiently, and absorb within the therapeutic window (600-850 nm). Traditional prediction methods for these light absorption properties, including Time-Dependent Density Functional Theory (TDDFT), are often computationally intensive and time-consuming. In this study, we explore a machine learning (ML) approach to predict the light absorption in the region of the therapeutic window of platinum, iridium, ruthenium, and rhodium complexes, aiming at streamlining the screening of potential photoactivatable prodrugs. By compiling a dataset of 9775 complexes from the Reaxys database, we trained six classification models, including random forests, support vector machines, and neural networks, utilizing various molecular descriptors. Our findings indicate that the Extreme Gradient Boosting Classifier (XGBC) paired with AtomPairs2D descriptors delivers the highest predictive accuracy and robustness. This ML-based method significantly accelerates the identification of suitable compounds, providing a valuable tool for the early-stage design and development of phototherapy drugs. The method also allows to change relevant structural characteristics of a base molecule using information from the supervised approach.Scientific Contribution: The proposed machine learning (ML) approach predicts the ability of transition metal-based complexes to absorb light in the UV-vis therapeutic window, a key trait for phototherapeutic agents. While ML models have been used to predict UV-vis properties of organic molecules, applying this to metal complexes is novel. The model is efficient, fast, and resource-light, using decision tree-based algorithms that provide interpretable results. This interpretability helps to understand classification rules and facilitates targeted structural modifications to convert inactive complexes into potentially active ones.
Collapse
Affiliation(s)
- V Vigna
- PROMOCS Laboratory, Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende (CS), Italy.
| | - T F G G Cova
- Coimbra Chemistry Centre, Department of Chemistry, Institute of Molecular Sciences (IMS), Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - A A C C Pais
- Coimbra Chemistry Centre, Department of Chemistry, Institute of Molecular Sciences (IMS), Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - E Sicilia
- PROMOCS Laboratory, Department of Chemistry and Chemical Technologies, University of Calabria, Arcavacata di Rende (CS), Italy
| |
Collapse
|
27
|
Anh Nguyen T, Thi Le P, Ho TH, Van Vu S, Nu Hoang Lo T, Park I, Pham NNT, Quoc Vo K. Synthesis of Urchin-Like Au@TiO 2 Nano-Carriers as a Drug-Loading System Toward Cancer Treatment. Chempluschem 2025; 90:e202400420. [PMID: 39331587 DOI: 10.1002/cplu.202400420] [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: 06/19/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
In recent years, improving the pharmaceutical properties of drug delivery for anti-cancer treatment has become increasingly important. This is necessary to address challenges related to absorption, distribution, and stability. One potential approach solution is to attach the drug to a carrier system, such as functional noble nanomaterials, in order to improve the control of drug release and stability. Core-satellite nanoparticles (CSN) with an anisotropic morphology have enormous potential for targeted drug delivery and cancer treatment because of their large surface area, exceptional stability, and biocompatibility. We used a simple seed-mediated approach to synthesize urchin-like gold nanoparticles (ULGNPs) with a high aspect ratio and a dense network of 49 nm-sized branches, using seed solution, silver nitrate, and ascorbic acid. The ULGNPs were synthesized without a surfactant and then encapsulated with thin layers of amorphous TiO2 (ULGNPs@TiO2), resulting in an average overall size of 136±15 nm with a 27.5 nm TiO2 layer. Doxorubicin (Dox) was chosen as a model drug to assess the distribution carrier ability of ULGNPs@TiO2 core-satellite nanoparticles. The results showed 86.5 % Dox loading and 72.3 % release capacity at pH 5. The anti-cancer ability of ULGNPs@TiO2-Dox was meticulously assessed using breast cancer MCF-7 cells in the WST-1 assay. The results revealed that ULGNPs@TiO2-Dox exhibited approximately 92 % toxicity in MCF-7 cells compared to the free Dox of 89.6 % at low concentrations (5 ppm). Based on the simulation results for loading ULGNPs@TiO2 with Dox, it was observed that a structure containing five layers of Au (111) with three fixed bottom layers and two relaxed top layers, in addition to six TiO2 (100) layers, was analyzed using Grimme's DFT-D3 dispersion corrections (Scheme 1). The density functional theory (DFT) adsorption energy (Eads) shows that the amorphous TiO2 increases the Dox loading activity of ULGNPs, with Eads=-3.85 eV, negatively higher than isolated ULGNPs (Eads=-2.87 eV) and TiO2 alone (Eads=-3.61 eV). This drug carrier design has the potential to revolutionize anti-cancer treatment.
Collapse
Affiliation(s)
- Thu Anh Nguyen
- Faculty of Chemistry, Ho Chi Minh City University of Science, Vietnam National University, Ho Chi Minh City, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, 70000, Vietnam
- Department of Physical Chemistry, Vietnam National University, Ho Chi Minh City, 70000, Vietnam
| | - Phuong Thi Le
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, 70000, Vietnam
| | - Thi H Ho
- Laboratory for Computational Physics, Institute for Computational Science and Artificial Intelligence, Van Lang University, Ho Chi Minh City, Vietnam
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, 70000, Vietnam
| | - Sy Van Vu
- Faculty of Chemistry, Ho Chi Minh City University of Science, Vietnam National University, Ho Chi Minh City, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, 70000, Vietnam
- Department of Physical Chemistry, Vietnam National University, Ho Chi Minh City, 70000, Vietnam
| | - Tien Nu Hoang Lo
- Research Institute of Clean Manufacturing System, Korea Institute of Industrial Technology (KITECH), 89 Yangdaegiro-gil, Ipjang-myeon, Cheonan, 31056, South Korea
- KITECH school, Department of Convergence Manufacturing System Engineering, University of Science and Technology (UST), 176 Gajeong-dong, Yuseong-gu, Daejeon, 34113, South Korea
| | - In Park
- Research Institute of Clean Manufacturing System, Korea Institute of Industrial Technology (KITECH), 89 Yangdaegiro-gil, Ipjang-myeon, Cheonan, 31056, South Korea
- KITECH school, Department of Convergence Manufacturing System Engineering, University of Science and Technology (UST), 176 Gajeong-dong, Yuseong-gu, Daejeon, 34113, South Korea
| | - Nguyet N T Pham
- Faculty of Chemistry, Ho Chi Minh City University of Science, Vietnam National University, Ho Chi Minh City, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, 70000, Vietnam
- Department of Physical Chemistry, Vietnam National University, Ho Chi Minh City, 70000, Vietnam
| | - Khuong Quoc Vo
- Faculty of Chemistry, Ho Chi Minh City University of Science, Vietnam National University, Ho Chi Minh City, 227 Nguyen Van Cu Street, Ward 4, District 5, Ho Chi Minh City, 70000, Vietnam
- Department of Physical Chemistry, Vietnam National University, Ho Chi Minh City, 70000, Vietnam
| |
Collapse
|
28
|
Song CW, Kim H, Kim MS, Park HJ, Paek SH, Terezakis S, Cho LC. Role of HIF-1α in the Responses of Tumors to Radiotherapy and Chemotherapy. Cancer Res Treat 2025; 57:1-10. [PMID: 38853541 PMCID: PMC11729307 DOI: 10.4143/crt.2024.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
Tumor microenvironment is intrinsically hypoxic with abundant hypoxia-inducible factors-1α (HIF-1α), a primary regulator of the cellular response to hypoxia and various stresses imposed on the tumor cells. HIF-1α increases radioresistance and chemoresistance by reducing DNA damage, increasing repair of DNA damage, enhancing glycolysis that increases antioxidant capacity of tumors cells, and promoting angiogenesis. In addition, HIF-1α markedly enhances drug efflux, leading to multidrug resistance. Radiotherapy and certain chemotherapy drugs evoke profound anti-tumor immunity by inducing immunologic cell death that release tumor-associated antigens together with numerous pro-immunological factors, leading to priming of cytotoxic CD8+ T cells and enhancing the cytotoxicity of macrophages and natural killer cells. Radiotherapy and chemotherapy of tumors significantly increase HIF-1α activity in tumor cells. Unfortunately, HIF-1α effectively promotes various immune suppressive pathways including secretion of immune suppressive cytokines, activation of myeloid-derived suppressor cells, activation of regulatory T cells, inhibition of T cells priming and activity, and upregulation of immune checkpoints. Consequently, the anti-tumor immunity elevated by radiotherapy and chemotherapy is counterbalanced or masked by the potent immune suppression promoted by HIF-1α. Effective inhibition of HIF-1α may significantly increase the efficacy of radiotherapy and chemotherapy by increasing radiosensitivity and chemosensitivity of tumor cells and also by upregulating anti-tumor immunity.
Collapse
Affiliation(s)
- Chang W Song
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hyunkyung Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Heon J Park
- Department of Microbiology, College of Medicine, Inha University, Incheon, Korea
| | - Sun-Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Stephanie Terezakis
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - L Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
29
|
Slly AM, Ewes WA, Bayoumi WA, Selim KB. Unveiling the potential anticancer activity of new dihydropyrimidines through dual inhibition of EGFR and TrkA: Design, synthesis, and in silico study. Bioorg Chem 2025; 154:107962. [PMID: 39591690 DOI: 10.1016/j.bioorg.2024.107962] [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: 10/10/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024]
Abstract
A series of designed scaffold of dihydropyrimidine was synthesized as dual tyrosine kinase targets inhibitors using a multicomponent Biginelli reaction which provided a high atom economy in a single pot reaction. Several 1,4-DHPM hybrids were obtained via alkylation with different chloroacetylamine derivatives. All the synthesized derivatives were screened for their antiproliferative efficacy towards various cancer cell lines (HCT-116, PC-3, and MCF-7) and normal cell line WI-38 using MTT assay. The results indicated that compounds 8h and 8i have the most significant inhibitory effect on all evaluated cancer cell lines, displaying IC50 of 3.94-15.78 µM. Also, they demonstrated favorable selectivity towards normal cell lines. Moreover, the most active hybrids 8h and 8i were evaluated for their EGFR and TrkA inhibitory activity. The findings indicated that compound 8h had superior inhibitory activity compared to compound 8i on the targeted kinases, effectively stopping the G1 phase of the MCF-7 cell cycle and encouraging apoptosis. Additionally, the molecular docking studies declared that the most active compounds exhibited a notable binding interaction with the binding site of the target proteins. Furthermore, their physicochemical properties, ADMET profiles, and bioavailability radar plots were predicted and analyzed.
Collapse
Affiliation(s)
- Aya M Slly
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Waleed A Bayoumi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Khalid B Selim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
30
|
Toruntay C, Poyraz FS, Susgun S, Yucesan E, Mansuroglu B. Anticancer Effects of MAPK6 siRNA-Loaded PLGA Nanoparticles in the Treatment of Breast Cancer. J Cell Mol Med 2025; 29:e70309. [PMID: 39823246 PMCID: PMC11740982 DOI: 10.1111/jcmm.70309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025] Open
Abstract
siRNA-loaded nanoparticles open new perspectives for cancer treatment. MAPK6 is upregulated in breast cancer and is involved in cell growth, differentiation and cell cycle regulation. Herein, we aimed to investigate the anticancer effects of MAPK6 knockdown by using MAPK6 siRNA-loaded PLGA nanoparticles (siMAPK6-PLGA-NPs) in MCF-7 breast cancer cells. After the synthesis and characterisation of nanoparticles, treatment concentrations were determined with cytotoxicity assay. Subsequently, MAPK6 knockdown and anticancer effects of siMAPK6-PLGA-NPs were evaluated by in vitro assays. siMAPK6-PLGA-NPs have been determined to suppress MAPK6 expression efficiently. In vitro studies revealed that siMAPK6-PLGA-NPs significantly reduced the migration, proliferation and colony-forming ability and enhanced the apoptosis in MCF-7 cells. Taken together, siMAPK6-PLGA-NPs exhibited robust and promising anticancer effects against MCF-7 cells. Our findings demonstrated that siRNA-loaded PLGA nanoparticles have great potential for breast cancer treatment and MAPK6 gene may be the therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Ceyhun Toruntay
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Turkiye
- Department of Molecular Biology and Genetics, Graduate School of Science and Engineering, Yildiz Technical University, Istanbul, Turkiye
| | - Fatma Sayan Poyraz
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Yildiz Technical University, Istanbul, Turkiye
| | - Seda Susgun
- Department of Genetics, Institute of Health Sciences, Istanbul University, Istanbul, Turkiye
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkiye
| | - Emrah Yucesan
- Department of Neurogenetics, Institute of Neurological Sciences, Istanbul University-Cerrahpasa, Istanbul, Turkiye
| | - Banu Mansuroglu
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Yildiz Technical University, Istanbul, Turkiye
| |
Collapse
|
31
|
Prajapati M, Deshmukh R, Harwansh RK. Recent Trends in Nanoparticulate Delivery System for Amygdalin as Potential Therapeutic Herbal Bioactive Agent for Cancer Treatment. Curr Drug Deliv 2025; 22:63-79. [PMID: 38037911 DOI: 10.2174/0115672018280381231119150732] [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: 09/02/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Cancer is the deadliest and most serious health problem. The mortality rate of cancer patients has increased significantly worldwide in recent years. There are several treatments available, but these treatments have many limitations, such as non-specific targeting, toxicity, bioavailability, solubility, permeability problems, serious side effects, and a higher dose. Many people prefer phytomedicine because it has fewer side effects. However, amygdalin is a naturally occurring phytoconstituent. It has many harmful effects due to the cyanide group present in the chemical structure. Many scientists and researchers have given their thoughts associated with amygdalin and its toxicities. However, there is a need for a more advanced, effective, and newer delivery system with reduced toxicity effects of amygdalin. Nanotechnology has become a more refined and emerging medical approach, offering innovative research areas to treat cancer. This review focuses on the use of amygdaline as herbal medicine encapsulating into several nanoparticulate delivery systems such as silver nanoparticles, graphene oxide nanoparticles, gold nanoparticles, nanofibers, nanocomposites, niosomes, and magnetic nanoparticles in the treatment of cancer. In addition, this article provides information on amygdalin structure and physical properties, pharmacokinetics, toxicity, and challenges with amygdalin.
Collapse
Affiliation(s)
- Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| |
Collapse
|
32
|
Abdul Ghafoor N, Rasuli S, Tanriverdi Ö, Yildiz A. Investigating the P53-dependent anti-cancer effect of ibutamoren in human cancer cell lines. Basic Clin Pharmacol Toxicol 2025; 136:e14111. [PMID: 39668330 DOI: 10.1111/bcpt.14111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024]
Abstract
The MDM2-p53 pathway plays a pivotal role in regulating cell cycle and apoptosis, with its dysfunction contributing to approximately 50% of human malignancies. MDM2, an E3 ubiquitin ligase, targets the tumour suppressor p53 for degradation, thereby promoting uncontrolled cell growth in cancers. Inhibiting the MDM2-p53 interaction represents a promising therapeutic strategy for reactivating p53's tumour-suppressive functions. This study explored the potential of ibutamoren (IBU) as a novel inhibitor of MDM2. In silico analyses utilizing molecular modelling revealed that IBU has a low IC50 for MDM2 inhibition and favourably binds to the p53-binding pocket of MDM2. In vitro experiments demonstrated that IBU treatment reduced the viability of immortalized cancer cell lines with a functional MDM2-p53 pathway but not in cell lines where this pathway harboured damaging mutations. This trend was further supported by RT-qPCR analysis, which showed differential expression of two p53 target genes upon IBU treatment in cell lines with wild MDM2-p53 pathways but not in those harbouring damaging mutations. These findings provide preliminary evidence supporting IBU's anticancer activity, plausibly through the MDM2-p53 pathway, and suggest that further studies are warranted to explore its mechanism of action and potential development as a lead compound in oncology research.
Collapse
Affiliation(s)
- Naeem Abdul Ghafoor
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Mugla Sitki Kocman University, Mugla, Turkey
| | - Sabina Rasuli
- UFR Biosciences, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Özgür Tanriverdi
- Department of Medical Oncology, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Ayşegül Yildiz
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Mugla Sitki Kocman University, Mugla, Turkey
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
33
|
Koothradan S, Nayeem S, Elyas KK. PEGylated iron oxide-gold core-shell nanoparticles for tumor-targeted delivery of Rapamycin. 3 Biotech 2025; 15:23. [PMID: 39735611 PMCID: PMC11669639 DOI: 10.1007/s13205-024-04189-y] [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: 09/09/2024] [Accepted: 12/07/2024] [Indexed: 12/31/2024] Open
Abstract
Rapamycin analogs are approved by the FDA for breast and renal cancer treatment. Hence, the possibility of nanoparticle-mediated delivery of Rapamycin could be examined. In the present study, PEGylated Gold-core shell iron oxide nanoparticles were used for the targeted delivery of Rapamycin, and R-Au-IONPs were formulated. SEM, XRD, and FTIR determined the smooth spherical morphology, and compositional structure, and confirmed the conjugation of Rapamycin onto the NPs. The in vitro drug release study showed a controlled release of the drug over time. R-Au-IONPs showed significant cytotoxicity in MCF 7 cells. Anti-proliferative assays such as trypan blue dye exclusion assay, microscopy, Fluorescent staining, and clonogenic assays were performed. NH staining, Rhodamine 123 staining, PS externalization, and the cleavage of PARP protein by western immunoblot assays confirmed the induction of apoptosis. The mechanism of R-Au-IONP-induced cell death was analyzed by flow cytometry. Our in-vitro study, on the impact of R-Au-IONPs on cell viability in the human breast adenocarcinoma cell line (MCF-7), confirms the efficacy of drug delivery using the nanoparticle system. Further results implied the induction of apoptosis. This drug delivery system using Rapamycin could be a potential candidate in the treatment of breast cancer.
Collapse
Affiliation(s)
- Suhana Koothradan
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| | - Safia Nayeem
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| | - K. K. Elyas
- Department of Biotechnology, University of Calicut, Kerala Malappuram, 673635 India
| |
Collapse
|
34
|
Jing HH, Shati AA, Alfaifi MY, Elbehairi SEI, Sasidharan S. The future of plant based green carbon dots as cancer Nanomedicine: From current progress to future Perspectives and beyond. J Adv Res 2025; 67:133-159. [PMID: 38320729 PMCID: PMC11725112 DOI: 10.1016/j.jare.2024.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/18/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The emergence of carbon dots (CDs) as anticancer agents had sparked a transformation in cancer research and treatment strategies. These fluorescent CDs, initially introduced in the early 2000 s, possess exceptional biocompatibility, tunable fluorescence, and surface modification capabilities, positioning them as promising tools in biomedical applications. AIM OF REVIEW The review encapsulates the transformative trajectory of green CDs as future anticancer nanomedicine, poised to redefine the strategies employed in the ongoing fight against cancer. KEY SCIENTIFIC CONCEPTS OF REVIEW The versatility of CDs was rooted in their various synthesis approaches and sustainable strategies, enabling their adaptability for diverse therapeutic uses. In vitro studies had showcased CDs' selective cytotoxicity against cancer cells while sparing healthy counterparts, forming the basis for targeted therapeutic potential. This selectivity had been attributed to the reactive oxygen species (ROS) generation, which opened avenues for targeted interventions. The role of CDs in combination therapies, synergizing with chemotherapy, radiotherapy, and targeted approaches was then investigated to heighten their anticancer efficacy. Notably, in vivo studies highlight CDs' remarkable biocompatibility and minimal side effects, endorsing their translational promise. Integration with conventional cancer treatments such as chemotherapy, radiotherapy, and immunotherapy amplified the versatility and effectiveness of CDs. The exploration of CDs' applications in photo-induced treatments further solidified their significance, positioning them as photosensitizers (PS) in photodynamic therapy (PDT) and photothermal agents (PA) in photothermal therapy (PTT). In PDT, CDs triggered the generation of ROS upon light exposure, facilitating cancer cell elimination, while in PTT, they induced localized hyperthermia within cancer cells, enhancing therapeutic outcomes. In vitro and in vivo investigations validated CDs' efficacy in PDT and PTT, affirming their potential for integration into combination therapies. Looking ahead, the future of CDs in anticancer treatment encompasses bioavailability, biocompatibility, synergistic treatments, tumor targeting, artificial intelligence (AI) and robotics integration, personalized medicine, and clinical translation. This transformative odyssey of CDs as future anticancer agents is poised to redefine the paradigm of cancer treatment strategies.
Collapse
Affiliation(s)
- Hong Hui Jing
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Pulau Pinang 11800, Malaysia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Pulau Pinang 11800, Malaysia.
| |
Collapse
|
35
|
Henrique RBL, Lima JVA, Santos ALF, Souza THS, Santos BS, Cabral Filho PE, Fontes A. Quantum dot-based conjugates: Luminous nanotools for cancer research. Biochem Biophys Res Commun 2024; 741:151052. [PMID: 39612641 DOI: 10.1016/j.bbrc.2024.151052] [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: 07/07/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
Nanotechnology has opened a new era in life science research, offering innovative nanotools to understand complex biological processes, such as those associated with cancer. Among the nanosystems, quantum dots (QDs) stand out for their remarkable optical properties, which render them valuable fluorescent nanoprobes in biological investigations. Moreover, their chemically active surfaces allow conjugations with (bio)molecules, other nanoparticles (NPs), and electrodes, enabling their use in multimodal site-specific applications and biosensing. The complexity and heterogeneity of cancer present challenges for its early diagnosis and personalized treatments. The conjugation of QDs with biologically relevant molecules can provide versatile nanotools for untangling cell mechanisms and biomarker patterns, thereby advancing the knowledge of cancer biology. This review illustrates the multifaceted capabilities of QDs, particularly in cancer research, drawing from applications at cell and tissue levels involving their conjugation with (i) low molecular weight molecules (e.g., folic acid, boronic acid, and glucose analog), (ii) macromolecules (e.g., holo-transferrin, lectins, and protease inhibitor), and (iii) optical-magnetic nanosystems combining QDs with superparamagnetic iron oxide NPs and holo-transferrin. The review also brings an overview of the fundamentals of QDs and strategies for their conjugation. By synthesizing findings from a range of studies, we hope that this review inspires new applications of QD-based conjugates in cancer biology, gathering knowledge and contributing to developing enhanced diagnostic and therapeutic procedures for this disease.
Collapse
Affiliation(s)
- Rafaella B L Henrique
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - João V A Lima
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Ana L F Santos
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Tiago H S Souza
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Beate S Santos
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, Pernambuco, 50740-520, Brazil
| | - Paulo E Cabral Filho
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Adriana Fontes
- Biomedical Nanotechnology Research Group (NanoBio), Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil.
| |
Collapse
|
36
|
Ma L, Kim MO. Advances in Preventive and Therapeutic Strategies for Oral Cancer: A Short Review. J Cancer Prev 2024; 29:113-119. [PMID: 39790224 PMCID: PMC11706729 DOI: 10.15430/jcp.24.027] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Oral cancer is a major global health concern, with high incidence and mortality rates, especially in high-risk populations. Early diagnosis remains a challenge, and current treatments, such as surgery, radiation, and chemotherapy, have limited effectiveness, particularly in advanced stages. Recent advances in targeted therapies and immunotherapy offer promising alternatives, providing more precise and personalized treatment options. Targeted therapies, such as epidermal growth factor receptor inhibitors, aim to disrupt specific molecular pathways in tumor growth, while immunotherapies, including immune checkpoint inhibitors and chimeric antigen receptor-T cell therapy, enhance the body's immune response to fight cancer. Combination therapies, integrating both targeted and immune strategies, are being explored to overcome the limitations of single-agent treatments. This review highlights the current strategies in the prevention and treatment of oral cancer, discusses emerging therapies, explores future research directions, focusing on optimizing existing treatments, identifying new biomarkers, and developing innovative therapeutic approaches. The potential of personalized medicine and combination therapies offers new hope for improving survival rates and quality of life for oral cancer patients.
Collapse
Affiliation(s)
- Lei Ma
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| |
Collapse
|
37
|
Klg A, Priyadharshini B, Vasugi S, Dilipan E. Exploring the therapeutic potential of biosynthetic enzymes in cancer treatment: Innovations and implications. Int J Biol Macromol 2024; 292:139171. [PMID: 39732247 DOI: 10.1016/j.ijbiomac.2024.139171] [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: 09/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Cancer remains a major global health concern due to several factors. These include the difficulty in accessing effective drugs, the high toxicity of available treatments, and the emergence of resistance to therapy. As a result, alternative strategies, such as the use of microbial enzymes, have gained attention as potential solutions to these challenges. Microbial enzymes have shown promise in inhibiting the uncontrolled growth of tumor cells through various mechanisms. In this comprehensive review, our objective is to emphasize the importance of pivotal microbial enzymes in fighting cancer and their ability to hinder the growth of tumors or cancer cells. The review article serves as a scientific roadmap for researchers, clinicians, and industry stakeholders exploring the therapeutic potential of biosynthetic enzymes in cancer treatment. It emphasizes the quest for effective and sustainable cancer therapies, presenting the possibility of personalized treatments with fewer side effects than traditional therapies.
Collapse
Affiliation(s)
- Afeeza Klg
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Boopathy Priyadharshini
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Suresh Vasugi
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Elangovan Dilipan
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
38
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
39
|
Amani AM, Tayebi L, Vafa E, Bazargan-Lari R, Abbasi M, Vaez A, Kamyab H, Gnanasekaran L, Chelliapan S, Azizli MJ. Innovative cancer therapy: Unleashing the potential of macromolecule-loaded mesoporous bioactive glasses for precision diagnosis and treatment. Int J Pharm 2024; 667:124847. [PMID: 39486491 DOI: 10.1016/j.ijpharm.2024.124847] [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: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024]
Abstract
Cancer continues to pose a formidable threat, claiming millions of lives annually. A beacon of hope in this battle lies in the realm of bioactive glasses, which have undergone a remarkable evolution over the past five decades. Among these, mesoporous bioactive glasses (MBGs) emerge as a dynamic subset endowed with customizable attributes such as high surface area and porosity. While holding immense promise for cancer care, the full clinical potential of MBGs remains largely unexplored. This review delves into the cutting-edge advancements in MBG technology, illuminating their pivotal role in cancer management - spanning from early detection to targeted therapeutic interventions like photothermal and photodynamic treatments. Furthermore, the molecular mechanisms underpinning MBGs' anticancer properties are elucidated, alongside an exploration of existing limitations in their application. Through this comprehensive synthesis, the significance of MBGs in revolutionizing cancer therapy is underscored, underscoring the urgent need for continued research to unlock their full potential in reshaping the landscape of cancer care.
Collapse
Affiliation(s)
- Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Institute for Engineering in Medicine, Health & Human Performance (EnMed), Batten College of Engineering and Technology, Old Dominion University, Norfolk, VA 23529, USA
| | - Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Bazargan-Lari
- Department of Materials Science and Engineering, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Universidad UTE, Centro de Investigación en Salud Públicay Epidemiología Clínica (CISPEC), Quito 170527, Ecuador; Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India; The KU-KIST Graduate School of Energy and Environment, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea.
| | | | - Shreeshivadasan Chelliapan
- Department of Smart Engineering and Advanced Technology, Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Mohammad Javad Azizli
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
40
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
41
|
Chekaoui A, Garofalo M, Gad B, Staniszewska M, Chiaro J, Pancer K, Gryciuk A, Cerullo V, Salmaso S, Caliceti P, Masny A, Wieczorek M, Pesonen S, Kuryk L. Cancer vaccines: an update on recent achievements and prospects for cancer therapy. Clin Exp Med 2024; 25:24. [PMID: 39720956 DOI: 10.1007/s10238-024-01541-7] [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: 08/26/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Decades of basic and translational research have led to a momentum shift in dissecting the relationship between immune cells and cancer. This culminated in the emergence of breakthrough immunotherapies that paved the way for oncologists to manage certain hard-to-treat cancers. The application of high-throughput techniques of genomics, transcriptomics, and proteomics was conclusive in making and expediting the manufacturing process of cancer vaccines. Using the latest research technologies has also enabled scientists to interpret complex and multiomics data of the tumour mutanome, thus identifying new tumour-specific antigens to design new generations of cancer vaccines with high specificity and long-term efficacy. Furthermore, combinatorial regimens of cancer vaccines with immune checkpoint inhibitors have offered new therapeutic approaches and demonstrated impressive efficacy in cancer patients over the last few years. In the present review, we summarize the current state of cancer vaccines, including their potential therapeutic effects and the limitations that hinder their effectiveness. We highlight the current efforts to mitigate these limitations and highlight ongoing clinical trials. Finally, a special focus will be given to the latest milestones expected to transform the landscape of cancer therapy and nurture hope among cancer patients.
Collapse
Affiliation(s)
- Arezki Chekaoui
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
| | - Beata Gad
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Jacopo Chiaro
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Aleksander Gryciuk
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Vincenzo Cerullo
- Drug Research Program (DRP), ImmunoViroTherapy Lab (IVT), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki, Helsinki, Finland
- Translational Immunology Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Digital Precision Cancer Medicine Flagship (iCAN), University of Helsinki, Helsinki, Finland
- Department of Molecular Medicine and Medical Biotechnology and CEINGE, University Federico II of Naples, Naples, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Aleksander Masny
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | | | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland.
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy.
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland.
- Valo Therapeutics Oy, Helsinki, Finland.
| |
Collapse
|
42
|
Goh CC, Gan XM, Klainin-Yobas P. Effectiveness of Digital-Based Interventions on Physical and Psychological Outcomes Among Cancer Patients: A Systematic Review and Meta-Analysis. Semin Oncol Nurs 2024:151796. [PMID: 39721898 DOI: 10.1016/j.soncn.2024.151796] [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: 08/30/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Evolving digital technology has paved the way for endless potentiality. Leveraging on digital technology for healthcare purposes can target cancer patients, thus improving physical and psychological symptoms. Nevertheless, there is limited consolidated evidence on the effectiveness of virtual reality (VR) and mobile applications. This review aimed to synthesize evidence concerning the effectiveness of VR and mobile-based interventions on physical (pain, fatigue, and sleep) and psychological (anxiety and depression) outcomes among cancer patients. METHODS A comprehensive literature search was conducted on ten electronic databases, ongoing trials, and grey literature, reported between 2013 to 2023. All randomized controlled trials (RCTs) examining VR and mobile-based interventions on the physical or psychological outcomes among people with cancer were included. Two independent reviewers screened records for eligibility, appraised methodological quality, and extracted data from included studies. The Cochrane Risk of Bias tool was used for data appraisal, and a modified Cochrane data extraction form was used for data extraction. Meta-analysis and subgroup analysis were used to analyze data. RESULTS In total, 43 studies were included. VR relaxation videos and game-based activities were efficacious in improving cancer-related pain. Mobile applications with educational content, symptom monitoring, relaxation videos, teleconsultation, and regular reminders contributed to positive effects on all outcomes. Mindfulness practice appeared to improve sleep quality, anxiety, and depression. Intervention effects sustained at least 6 months for all outcomes, except sleep. CONCLUSIONS VR and mobile-based interventions had the potential to improve pain, fatigue, sleep, anxiety, and depression at post-intervention. Future RCTs are required to further test both digital interventions on specific types of cancer on multiple research settings. IMPLICATIONS TO NURSING PRACTICE VR and mobile-based interventions can be offered in clinical settings to help cancer manage their pain, fatigue, sleep, anxiety, and depression. VR relaxation videos, game-based activities, teleconferences, mindfulness, education, and system reminders can be included.
Collapse
Affiliation(s)
| | - Xi Meng Gan
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Piyanee Klainin-Yobas
- Associate Professor, Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
43
|
Peng Y, Yang X, Liu Y, Zhou J, Guo J, Ma B, Bai Y, Wu J, Hu D. The regulation of the cell cycle and epithelial-mesenchymal transition through FUCA2/GGH signaling promotes the progression of lung adenocarcinoma. Gene 2024; 939:149183. [PMID: 39710011 DOI: 10.1016/j.gene.2024.149183] [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: 09/25/2024] [Revised: 11/14/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The development of lung adenocarcinoma (LUAD) is intricately linked with cell cycle regulation and epithelial-mesenchymal transition (EMT). Our study, leveraging bioinformatics and database analysis, identified FUCA2 as a key gene influencing the prognosis and progression of LUAD. We observed that FUCA2 is highly expressed in LUAD and correlates with poor outcomes. Functionally, we assessed the role of this gene through cell cloning, scratch assays, transwell migration, and western blotting, revealing that FUCA2 knockdown significantly inhibits tumor cell proliferation and migration, downregulates the expression of cell cycle and EMT-related proteins, and markedly reduces tumor burden. Mechanistically, pathway enrichment analysis identified GGH as a downstream target of FUCA2. Knockdown of GGH similarly inhibits the proliferation, migration, and cell cycle progression of LUAD cells. FUCA2 upregulates GGH to modulate cell cycle and EMT in LUAD. Collectively, our findings indicate that the FUCA2/GGH axis promotes LUAD progression by regulating cell cycle and EMT.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Xingyu Yang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yafeng Liu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jiawei Zhou
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jianqiang Guo
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Bingfeng Ma
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Ying Bai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Dong Hu
- The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei City, China.
| |
Collapse
|
44
|
Louis D, Rizkalla CMZ, Rashad A. Cubosomes as Delivery System to Repositioning Nitrofurantoin in Breast Cancer Management. Drug Des Devel Ther 2024; 18:6173-6184. [PMID: 39722678 PMCID: PMC11668685 DOI: 10.2147/dddt.s499068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose Nitrofurantoin (NITRO), a long-standing antibiotic to treat urinary tract infections, is activated by Nitro reductases. This activation mechanism has led to its exploration for repositioning applications in controlling and treating breast cancer, which express a Nitro reductase gene. Methods NITRO Cubosomes were developed using hot homogenization according to 23-full factorial design. The factors studied included the ratio of drug to oily phase (1:10 and 2:10), the ratio of oily to aqueous phase (1:10 and 1:5), and the ratio of Glyceryl mono-oleate (GMO) to Poloxamer 407 (PX407) (0.25:1 and 0.5:1). A total of 8 systems were proposed and evaluated by measuring particle size, zeta potential, polydispersity index, and percentage of entrapment efficiency. Results S6 (1:10 drug: oily phase, 1:5 oily: aqueous phase and 0.5:1 GMO: PX407) with particle size 45.5 ±c1.1 nm and an entrapment efficiency of 98.6 ± 1.8% exhibited highest desirability and was selected for further analysis. The morphology of S6 was examined using TEM microscopy. The activation of NITRO from S6 reflected on intracellular viability of MCF-7 breast cancer cell line was investigated by an MTT assay. The findings indicated that S6 had the lowest IC50 value (83.99 ± 0.15 μg g/mL) compared to Free NITRO (174.54 ± 1.36 μg g/mL), suggesting enhanced efficacy compared to free NITRO. Conclusion Nitrofurantoin cubosomes can be candidates for repositioning in breast cancer management after encouraging further stability and in-vivo studies.
Collapse
Affiliation(s)
- Dina Louis
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | | | - Amira Rashad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
45
|
St-Laurent MP, Bochner B, Catto J, Davies BJ, Fankhauser CD, Garg T, Hamilton-Reeves J, Master V, Jensen BT, Lauridsen SV, Wulff-Burchfield E, Psutka SP. Increasing Life Expectancy in Patients with Genitourinary Malignancies: Impact of Treatment Burden on Disease Management and Quality of Life. Eur Urol 2024:S0302-2838(24)02746-5. [PMID: 39706786 DOI: 10.1016/j.eururo.2024.11.026] [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: 09/11/2024] [Revised: 10/24/2024] [Accepted: 11/24/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Treatment burden refers to the overall impact of medical treatments on a patient's well-being and daily life. Our objective is to evaluate the impact of treatment burden on quality of life (QoL) in patients with genitourinary (GU) malignancies, highlighting the importance of patient-reported outcomes (PROs) in clinical trials to inform treatment decisions and improve patient care. METHODS We conducted a narrative review of clinical trials focused on GU malignancy (prostate, bladder, and kidney) between January 2000 and June 2024, analyzing related PROs and findings regarding treatment burden. KEY FINDINGS AND LIMITATIONS Recent landmark clinical trials demonstrate significant improvements in overall survival across GU malignancies with novel therapies. However, the reporting of QoL outcomes in these trials is often inadequate, with many lacking comprehensive data or long-term impact. Current publications are increasingly evaluating treatment burden and its impact on patient well-being as a critical outcome, but most clinical trials to date have failed to assess treatment burden across key domains including financial, time and travel, and medication management. CONCLUSIONS AND CLINICAL IMPLICATIONS While advancements in treatment have extended longevity in patients with GU malignancies, the treatment burden associated with the receipt of novel agents and its implications for QoL remain inadequately uncharacterized.
Collapse
Affiliation(s)
- Marie-Pier St-Laurent
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Bernard Bochner
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James Catto
- Department of Urology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Benjamin J Davies
- Department of Urology Division of Health Services Research University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Tullika Garg
- Department of Urology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Viraj Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bente T Jensen
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Susanne V Lauridsen
- WHO-CC/Clinical Health Promotion Centre, the Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark; Center for Perioperative Optimization, Department of Surgery, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elizabeth Wulff-Burchfield
- Medical Oncology Division and Palliative Medicine Division, Department of Internal Medicine, University of Kansas School of Medicine, University of Kansas Cancer Center, The University of Kansas Health System, Kansas City, KS, USA
| | - Sarah P Psutka
- Department of Urology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
46
|
Yuan P, Zhang W, Yang M, Chen Y, Yang Z, Zhao Z, Zhang L. Near-infrared light-enhanced polydopamine-based multifunctional nanoparticles for combination of chemodynamic and NO gas therapy in the treatment of osteosarcoma. Int J Biol Macromol 2024; 289:138946. [PMID: 39706438 DOI: 10.1016/j.ijbiomac.2024.138946] [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: 06/07/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
The emergence of treatment approaches that integrate conventional phototherapy with additional adjuvant treatments has garnered considerable interest. In this study, we proposed a complex utilizing Fe and polydopamine as a carrier, co-loaded with the nitric oxide initiator L-arginine (L-Arg) and the photosensitizer indocyanine green (ICG), as a potential strategy for the "photothermal/photodynamic/Chemodynamic/nitric oxide gas therapy" of osteosarcoma. Nanoparticles have the ability to undergo degradation within the mildly acidic conditions present in the tumor microenvironment. Consequently, the resulting release of Fe ions facilitates the consumption of hydrogen peroxide through Fenton/Fenton-like reactions, thereby generating hydroxyl radicals (•OH) that possess potent cytotoxic properties. L-Arg can also be catalyzed by reactive oxygen species (ROS) or NO synthase overexpressed in cancer cells to generate NO, which is not only used for gas therapy (GT), but also as a biological messenger to regulate vasodilation to relieve tumor hypoxia. More importantly, the addition of low-dose near-infrared laser can not only promote the efficiency of the above two reactions, but also achieve PTT/PDT and obtain good synergistic tumor treatment effects. The anti-tumor efficacy of nanoparticles was verified in the 143B mouse osteosarcoma model. This "PTT/PDT/CDT/GT" strategy expands bone tumor treatment options through nanoparticle-mediated enhanced therapy.
Collapse
Affiliation(s)
- Ping Yuan
- Sanya Science and Education Innovation Park, Wuhan University of Technology, Sanya 572000, China; State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China.
| | - Weigang Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Mingrui Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Yun Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Zhifei Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Zheng Zhao
- Sanya Science and Education Innovation Park, Wuhan University of Technology, Sanya 572000, China; State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China.
| | - Lingling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
47
|
Ravichandiran P, Martyna A, Kochanowicz E, Maroli N, Kubiński K, Masłyk M, Boguszewska-Czubara A, Ramesh T. In Vitro and In Vivo Biological Evaluation of Novel 1,4-Naphthoquinone Derivatives as Potential Anticancer Agents. ChemMedChem 2024; 19:e202400495. [PMID: 39136593 DOI: 10.1002/cmdc.202400495] [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: 06/29/2024] [Revised: 08/13/2024] [Indexed: 10/16/2024]
Abstract
A novel library of naphthoquinone derivatives (3-5 aa) was synthesized and evaluated for their anticancer properties. Specifically, compounds 5 i, 5 l, 5 o, 5 q, 5 r, 5 s, 5 t, and 5 v demonstrated superior cytotoxic activity against the cancer cell lines that were studied. All the studied compounds exhibited a higher selectivity index (SI) and a favourable safety profile than the standard drug doxorubicin. Notably, compound 5 v displayed a greater cytotoxic effect on MCF-7 cells (IC50=1.2 μM, and 0.9 μM at 24 h and 48 h, respectively) compared to the standard drug doxorubicin (IC50=2.4 μM, and 2.1 μM at 24 h and 48 h, respectively). To further investigate the mechanism of cytotoxic effect, additional anticancer studies were conducted with 5 v in MCF-7 cells. The studies are including morphological changes, AO/EB (acridine orange/ethidium bromide) double staining, apoptosis analysis, cell colony assay, SDS-PAGE and Western blotting, cell cycle analysis, and detecting reactive oxygen species (ROS) assay. The findings showed that 5 v triggered cytotoxic effects in MCF-7 cells through the initiation of cell cycle arrest at the G1/S phase and necrosis. In vivo ecotoxicity studies indicated that 5 v had lower toxicity towards zebrafish larvae (LC50=50.15 μM) and had an insignificant impact on cardiac functions. In vivo xenotransplantation of MCF-7 cells in zebrafish larvae demonstrated a significant reduction in tumour volume in the xenograft. Approximately 95 % of the zebrafish larvae with 5 v xenografts survived after 10 days of the treatment. Finally, a computational modelling study was conducted on four protein receptors, namely ER, EFGR, BRCA1, and VEFGR2. The findings highlight the importance of the aminonaphthoquinone moiety, amide linkage, and propyl thio moiety in enhancing the anticancer properties. 5 v exhibited superior drug-likeness features and docking scores (-9.1, -7.1, -8.9, and -10.9 kcal/mol) compared to doxorubicin (-7.2, -6.1, -6.9, and -7.3 kcal/mol) against ER, EFGR, BRCA1, and VEGFR2 receptors, respectively. Therefore, the notable antitumor effects of naphthoquinone derivatives (3-5 aa) suggest that these molecular frameworks may play a role in the development of promising anticancer agents for cancer treatment.
Collapse
Affiliation(s)
- Palanisamy Ravichandiran
- R&D Education Center for Whole Life Cycle R&D of Fuel Cell Systems, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Department of Energy Storage/Conversion Engineering of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Present Address: Analytical, HP Green R & D Centre, Hindustan Petroleum Corporation Limited, KIADB Industrial Area, Devangundi, Hoskote, Bengaluru, Karnataka, 562114, India
| | - Aleksandra Martyna
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Elżbieta Kochanowicz
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Nikhil Maroli
- Department of Physics and Astronomy, University of Delaware, Newark, DE, 19716, USA
| | - Konrad Kubiński
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Ul. Chodźki 4 A, 20-093, Lublin, Poland
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
48
|
Maldonado-García JL, Fragozo A, Pavón L. Cytokine release syndrome induced by anti-programmed death-1 treatment in a psoriasis patient: A dark side of immune checkpoint inhibitors. World J Clin Cases 2024; 12:6782-6790. [PMID: 39687650 PMCID: PMC11525914 DOI: 10.12998/wjcc.v12.i35.6782] [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: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/24/2024] Open
Abstract
In recent years, cancer immunotherapy has introduced novel treatments, such as monoclonal antibodies, which have facilitated targeted therapies against tumor cells. Programmed death-1 (PD-1) is an immune checkpoint expressed in T cells that regulates the immune system's activity to prevent over-activation and tissue damage caused by inflammation. However, PD-1 is also expressed in tumor cells and functions as an immune evasion mechanism, making it a therapeutic target to enhance the immune response and eliminate tumor cells. Consequently, immune checkpoint inhibitors (ICIs) have emerged as an option for certain tumor types. Nevertheless, blocking immune checkpoints can lead to immune-related adverse events (irAEs), such as psoriasis and cytokine release syndrome (CRS), as exemplified in the clinical case presented by Zhou et al involving a patient with advanced gastric cancer who received sintilimab, a monoclonal antibody targeting PD-1. Subsequently, the patient experienced exacerbation of psoriasis and CRS. The objective of this editorial article is to elucidate potential immunologic mechanisms that may contribute to the development of CRS and psoriasis in patients receiving ICIs. It is crucial to acknowledge that while ICIs offer superior safety and efficacy compared to conventional therapies, they can also manifest irAEs affecting the skin, gastrointestinal tract, or respiratory system. In severe cases, these irAEs can lead to life-threatening complications such as circulatory shock or multiorgan failure. Consequently, it is recommended that patients receiving ICIs undergo regular monitoring to identify and manage these adverse events effectively.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán 04510, Ciudad de México, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Mexico City 1134, Ciudad de México, Mexico
| | - Ana Fragozo
- Unidad de Desarrollo e Investigación en Bioterapéuticos, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Ciudad de México, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 11340, Mexico
| |
Collapse
|
49
|
Madikonda AK, Ajayakumar A, Nadendla S, Banothu J, Muripiti V. Esterase-responsive nanoparticles (ERN): A targeted approach for drug/gene delivery exploits. Bioorg Med Chem 2024; 116:118001. [PMID: 39556942 DOI: 10.1016/j.bmc.2024.118001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Nanoparticles are being developed to enhance drug delivery to cancer tumors, leveraging advantages such as the enhanced permeability and retention (EPR) effect. However, traditional nanoparticles often face challenges with low specificity for cancer cells, leading to inefficient delivery and unwanted side effects. Esterase-responsive nanoparticles offer a maximum targeted approach to tumor cells because they release their therapeutic payload at the tumor site under the influence of esterase activity. This review explores the role of esterase-responsive nanoparticles in drug and gene delivery, examines esterase prodrug therapy, and discusses prostate-specific membrane antigen (PSMA) targets esterase-responsive nanoparticles in prostate cancer treatment. Additionally, we reviewed the current research progress and future potential of esterase-responsive nanoparticles in enhancing drug and gene delivery.
Collapse
Affiliation(s)
- Ashok Kumar Madikonda
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Amritha Ajayakumar
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Sudeena Nadendla
- Department of Chemistry, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Janardhan Banothu
- Department of Chemistry, National Institute of Technology Calicut, Kozhikode 673601, Kerala, India
| | - Venkanna Muripiti
- Department of Education, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India.
| |
Collapse
|
50
|
Khaled NA, Ibrahim MA, Mohamed NA, Ahmed SA, Ahmed NS. DFT studies on N-(1-(2-bromobenzoyl)-4-cyano-1H-pyrazol-5-yl). SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 323:124864. [PMID: 39067358 DOI: 10.1016/j.saa.2024.124864] [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: 04/30/2024] [Revised: 07/01/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
In this work, molecular descriptors of N-(1-(2-bromobenzoyl)-4-cyano-1H-pyrazol-5-yl) halogenated benzamides (1a-h) have been computed using a quantum chemical technique through DFT. Prior work involved the synthesis of compounds (1a-h) and the assessment of their anticancer activity on breast, colon, and liver tumors: MCF-7, HCT-116, and HepG-2 cell lines respectively. Since 1a, 1b, and 1d showed the most potential anticancer impact, their ability to inhibit EGFRWT was investigated. Based on the biological data, 1b inhibited EGFRWT the most. According to the docking evaluation, an H-bond with the threonine residue was one of the main non-covalent contacts between 1b and the EGFRWT active site residues. PES, MESP, HOMOs, LUMOs, energy band gap, global reactivity indices [electron affinity (A), ionization energies (I), electrophilicity index (ω), nucleophilicity index (ε), chemical potential (μ), electronegativity (χ), hardness (η), and softness (S)], condensed Fukui functions, NBO, and NCIs are the molecular descriptors of 1a-h that were computed using DFT technique. According to the theoretical investigation results, compounds (1a-h) might have anticancer effects; these findings are consistent with the biological findings from our previous research. Compound 1b had the lowest binding energy, according to an assessment of the binding energies between the threonine and the three most active compounds (1a, 1b, and 1d). This is consistent with the outcomes of the docking study and the biological examination of the influence of 1a, 1b, and 1d on EGFRWT.
Collapse
Affiliation(s)
- Nada A Khaled
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Medhat A Ibrahim
- Spectroscopy Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt; Molecular Modeling and Spectroscopy Laboratory, Centre of Excellence for Advanced Science, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt.
| | - Neama A Mohamed
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Sayed A Ahmed
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt; Basic Science Department, Faculty of Engineering, Nahda University Beni-Suef (NUB), Beni Suef, Egypt
| | - Nesreen S Ahmed
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|