1
|
Jamal MH, Khan MN. Developments in pancreatic cancer emerging therapies, diagnostic methods, and epidemiology. Pathol Res Pract 2025; 271:156012. [PMID: 40414076 DOI: 10.1016/j.prp.2025.156012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is still one of the deadliest malignancies, characterised by late-stage diagnosis, aggressive biology, and considerable resistance to conventional treatments. Despite improvements in understanding the molecular mechanisms and innovations in treatment, the overall survival remains abysmal: fewer than 9 % of patients survive beyond 5 years. By 2030, PC is predicted to become the second leading cause of cancer-related deaths in the U.S. owing to chemoresistance, rapid metastatic spread, and limited effective immunotherapeutic choices. This review highlights current progress in this field, including epidemiology, risk factors, diagnostic tools, and emerging biomarkers. Recent progress in genetic and molecular profiling has provided important information about pancreatic cancer. It has identified key mutations in genes like KRAS, TP53, CDKN2A, and SMAD4 that play a major role in driving the disease. Such revelations have provided the impetus to explore novel targeted therapies against these mutations. Furthermore, the advances in liquid biopsies incorporating circulating tumour cells, circulating tumour DNA, and exosomes hold substantial promise for early diagnosis, treatment response monitoring, and detection of minimal residual disease-any of which could radically transform PC management. While very limited options for the treatment of advanced-stage PC remain, the only potential curative treatment is surgery, yet only 10-15 % of patients are diagnosed with potentially resectable disease. Researchers are looking into new methods to help more patients qualify for surgery. This involves using chemotherapy and radiotherapy to reduce the size of the tumor before the operation. New chemotherapy treatments like FOLFIRINOX (which includes 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) have improved results for some patients, but they can still cause significant side effects. Immunotherapy, though revolutionary in other cancers, has had limited success in PC due to the tumour's immunosuppressive microenvironment. Researchers are looking into using immune checkpoint inhibitors together with chemotherapy, radiation, and drugs that target the surrounding tissue to improve the body's immune response. There is also considerable excitement surrounding personalised approaches with adoptive cell therapies such as CAR-T cells and TILs, which are trialled with early evidence of potential efficacy. Attempts are also being made to address the dense desmoplastic stroma of the tumour that characterises PC. Drugs that can fight resistance or new medicines that might affect the tumor environment, stop changes in surrounding tissues, and improve how drugs are delivered have shown some potential in laboratory tests so far. Nanoparticle-based drug delivery systems are also being developed to improve the bioavailability and targeted delivery of chemotherapy.
Collapse
Affiliation(s)
- Mohd Haris Jamal
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Md Nasiruddin Khan
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
2
|
Liu T, Zhang D, Ouyang W, Li R, Wang S, Liu W. Identification and functional characterization of hub genes CLTA, EDIL3, HAPLN1, and HIP1 as diagnostic biomarkers and therapeutic targets in thyroid cancer and Hashimoto's thyroiditis. Clin Exp Med 2025; 25:162. [PMID: 40372556 PMCID: PMC12081531 DOI: 10.1007/s10238-025-01689-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
In this study, we sought to identify key molecular players in both thyroid cancer (TC) and Hashimoto's thyroiditis (HT) by analyzing differentially expressed genes (DEGs) and their potential as biomarkers. We utilized datasets from the Gene Expression Omnibus (GEO) database and identified CLTA, EDIL3, HAPLN1, and HIP1 as hub genes common to both TC and HT. These genes were significantly upregulated in TC cell lines compared to normal controls, with high diagnostic accuracy as indicated by Receiver Operating Characteristic (ROC) curve analysis. Further validation using the TCGA TC dataset revealed their significant upregulation in tumor tissues, particularly in advanced TC stages. Promoter methylation analysis indicated hypomethylation of these genes in TC, suggesting a role of methylation in their regulation. We also observed mutations and copy number variations (CNVs) in these hub genes, with CLTA and HIP1 showing significant amplifications, which may contribute to their overexpression in tumor samples. In addition, we conducted a meta-analysis to assess the impact of these genes on survival outcomes in TC patients, with results indicating that higher expression of HAPLN1 and HIP1 was associated with poor survival. Our study also highlighted the involvement of CLTA and EDIL3 in activating the Rap1 signaling pathway, crucial for cancer cell migration, proliferation, and invasion. These findings emphasize the potential of CLTA, EDIL3, HAPLN1, and HIP1 as diagnostic biomarkers and therapeutic targets for TC and HT.
Collapse
Affiliation(s)
- Tianyu Liu
- Department of Geriatrics, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China
| | - Dechun Zhang
- Department of Geriatrics, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China
| | - Wen Ouyang
- Department of Medical Affairs, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China
| | - Rongfang Li
- Department of Geriatrics, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China
| | - Siying Wang
- Department of Geriatrics, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China
| | - Weixuan Liu
- Department of Geriatrics, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, 415000, China.
| |
Collapse
|
3
|
Raghani NR, Chorawala MR, Parekh K, Sharma A, Alsaidan OA, Alam P, Fareed M, Prajapati B. Exosomal miRNA-based theranostics in cervical cancer: bridging diagnostics and therapy. Med Oncol 2025; 42:193. [PMID: 40320487 DOI: 10.1007/s12032-025-02752-y] [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: 03/02/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025]
Abstract
Cervical cancer (CC) remains a significant global health burden, particularly in low- and middle-income countries, where access to effective screening and treatment is limited. Despite advancements in conventional therapies, such as surgery, chemotherapy, and radiotherapy, challenges related to late-stage diagnosis, treatment resistance, and disease recurrence persist. The emergence of microRNAs (miRNAs) as key regulators of gene expression has revolutionized cancer diagnostics and therapeutics. Exosomal miRNAs, in particular, have garnered attention due to their stability, detectability in bodily fluids, and pivotal roles in tumor progression, metastasis, and immune modulation. This review provides a comprehensive overview of the role of exosomal miRNAs in the theranostic landscape of CC. We explore their involvement in disease pathogenesis, highlighting their potential as minimally invasive diagnostic biomarkers for early detection and disease monitoring. Furthermore, we examine their utility in therapeutic strategies, including miRNA-mediated drug delivery systems and miRNA-targeted interventions to overcome chemoresistance. Integrating exosomal miRNA profiling with current diagnostic modalities could enhance screening sensitivity and specificity, while miRNA-based therapies offer novel avenues to improve treatment efficacy. This review discusses recent advancements in miRNA research, current challenges in clinical translation, and future perspectives on leveraging exosomal miRNAs for personalized CC care.
Collapse
Affiliation(s)
- Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India.
| | - Kavya Parekh
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Anvesha Sharma
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, 72341, Sakaka, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 13713, Saudi Arabia
| | - Bhupendra Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S K Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Mahesana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
4
|
Hussain SA, Ramasamy M, Shaik MR, Shaik B, Deepak P, Thiyagarajulu N, Matharasi Antonyraj AP, Guru A. Inhibition of Oral Biofilms and Enhancement of Anticancer Activity on Oral Squamous Carcinoma Cells Using Caffeine-Coated Titanium Oxide Nanoparticles. Chem Biodivers 2025; 22:e202402476. [PMID: 39715068 DOI: 10.1002/cbdv.202402476] [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: 10/01/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024]
Abstract
The fungus Candida albicans is a prominent cariogenic fungal agent that works in association with Streptococcus mutans to accelerate the formation of oral cancer and tooth decay. This study evaluates caffeine-encapsulated titanium oxide nanoparticles (CF-TiO2 NPs) for their potential to prevent biofilm formation on teeth and enhance oral anticancer treatment by influencing apoptotic gene regulation. The synthesized CF-TiO2 NPs were characterized using ultraviolet, scanning electron microscopy, energy-dispersive X-ray spectroscopy, and Fourier-transform infrared spectroscopy analyses, and their antioxidant activity was confirmed through free radical quenching studies. Antimicrobial efficacy was assessed using a zone of inhibition test, revealing strong activity against dental pathogens with a minimal inhibitory concentration of 80 µg/mL. Molecular docking using AutoDock explored interactions between CF and biofilm target sites, supporting their inhibitory potential. In vitro cytotoxicity studies on KB cancer cells showed a dose-dependent increase in cytotoxic effects, with CF-TiO2 NPs promoting apoptotic gene upregulation at concentrations of 20-160 µg/mL. CF-TiO2 NPs demonstrated excellent antioxidant, antibacterial, and anticancer properties, showcasing their promise for oral therapeutic applications. This research highlights a novel approach to managing oral infections and associated complications while improving systemic oral health. Notably, this study is the first to report the biofilm-inhibitory and anticancer potential of CF-TiO2 NPs.
Collapse
Affiliation(s)
- Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohankumar Ramasamy
- Department of Chemistry, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, India
- Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur, India
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Baji Shaik
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Paramasivam Deepak
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | | | - Anahas Perianaika Matharasi Antonyraj
- Department of Research Analytics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
5
|
Jafar NNA, Abd Hamid J, M A Altalbawy F, Sharma P, Kumar A, Shomurotova S, Jihad Albadr R, Atiyah Altameemi KK, Mahdi Saleh H, Alajeeli F, Mohammed Ahmed A, Ahmad I, Dawood II. Gadolinium (Gd)-based nanostructures as dual-armoured materials for microbial therapy and cancer theranostics. J Microencapsul 2025; 42:239-265. [PMID: 39992246 DOI: 10.1080/02652048.2025.2469259] [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: 08/31/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
Gadolinium (Gd) nanoparticles hold significant promise in medical theranostics due to their unique properties. This review outlines the synthesis, characterisation, and applications of Gd nanostructures in combating microbial threats and advancing cancer theragnostic strategies. Synthesis methods such as co-precipitation, microemulsion, and laser ablation are discussed, alongside TEM, SEM, and magnetic characterisation. The antimicrobial efficacy of Gd nanostructures, their potential in combination therapy, and promising anticancer mechanisms are explored. Biocompatibility, toxicity, and regulatory considerations are also evaluated. Challenges, future perspectives, and emerging trends in Gd nanostructure research are highlighted, emphasising their transformative potential in medical applications.
Collapse
Affiliation(s)
- Nadhir N A Jafar
- AL-Zahraa University for Women, College of Health and Medical Technology, Kerbala, Iraq
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Pawan Sharma
- Department of Chemistry, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Sciences, Vivekananda Global University, Jaipur, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, India
| | - Shirin Shomurotova
- Department of Chemistry Teaching Methods, Tashkent State Pedagogical University Named After Nizami, Tashkent, Uzbekistan
| | | | | | - Hawraa Mahdi Saleh
- Department of Dentistry, Al-Manara College For Medical Sciences, Maysan, Iraq
| | - Fakhri Alajeeli
- Department of Medical Laboratories Technology, Al-Hadi University College, Baghdad, Iraq
| | - Ahmed Mohammed Ahmed
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| | - Irfan Ahmad
- Central Labs, King Khalid University, AlQura'a, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Imad Ibrahim Dawood
- Department of Medical Laboratories Technology, Mazaya University College, Nasiriyah, Iraq
| |
Collapse
|
6
|
M A, S KB, Liyana EP, Jasmine JS. Transformative potential of plant-based nanoparticles in cancer diagnosis and treatment: bridging traditional medicine and modern therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04113-y. [PMID: 40237799 DOI: 10.1007/s00210-025-04113-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Cancer is a primary global health concern, with an estimated 35.3 million cancer cases expected worldwide, representing a 76.6% increase in 2022, and 20 million by 2050, resulting from genetic mutation and environmental factors that cause uncontrolled cell growth. Other factors including smoking, unhealthy diets, physical inactivity, exposure to carcinogens, UV radiation, and aging increase DNA damage. Current cancer treatments like chemotherapy, radiation therapy, immunotherapy, and surgery are effective, but those have significant effects like lack of specificity, development of drug resistance, and significant side effects to healthy tissues. An advancement to conventional therapies is plant-based nanoparticles as transformative approaches in cancer diagnosis and treatment. These nanoparticles synthesized using plant bioactive compounds like flavonoids, alkaloids, polyphenols, and some metals-oxides like gold, silver, copper, zinc, etc. offer eco-friendly, cost-effective, and biocompatible alternatives. They enhance targeted drug delivery, allowing anticancer agents specifically to tumor cells, minimizing damage to health. Improves imaging techniques like MRI and fluorescence imaging, and helps early detection, cancer biomarkers, allowing for prompt intervention. Recent findings show that nanocarriers made from plant-based materials, such as polyphenols (curcumin, resveratrol) and plant-extracted metal nanoparticles (gold, silver), can improve drug stability and selectively target tumor cells. Plant-derived nanoparticles play a crucial role in cancer immunotherapy and nanovaccines. Biodegradable plant-based nanocarriers can deliver cancer vaccines, stimulating long-term immunity against tumors. Graphene oxide and gold nanoparticles synthesized from plant extracts can absorb near-infrared (NIR) light, generating heat to destroy cancer cells with minimal damage to surrounding tissues. This study discusses the types of plant-based nanoparticles like plant virus nanoparticles (TMV, PVX, CPMV), plant metallic nanoparticles (Au, Ag., Cu, Zn, Mg, Ca, and Mn), and flavonoid nanoparticles found in cancer treatment, their significant roles, chemotherapy-based nanomedicines available in the medical field, and a detailed vision of nanomaterial applications in cancer diagnosis, treatment, and targeted drug delivery.
Collapse
Affiliation(s)
- Aswini M
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India.
| | - Kavitha Bagya S
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | - E P Liyana
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, Tamil Nadu, India
| | | |
Collapse
|
7
|
Bao J, Tian X, Pan Y, Guo Y, Yang Z, Gan M, Zheng J. SNRPB2: a prognostic biomarker and oncogenic driver in esophageal cancer via β-catenin/c-Myc signaling. Front Oncol 2025; 15:1536473. [PMID: 40303992 PMCID: PMC12037380 DOI: 10.3389/fonc.2025.1536473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Background The SNRPB2 gene encodes Small Nuclear Ribonucleoprotein Polypeptide B2, a crucial component involved in RNA splicing processes. While SNRPB2 dysregulation has been observed in various cancers, its role in esophageal cancer (ESCA) remains unclear. Methods The mRNA level of SNRPB2 in ESCA was evaluated in combination with TCGA, GTEX, and GEO databases. The prognostic value of SNRPB2 was assessed using Kaplan-Meier analysis. Immunohistochemistry (IHC) was employed to confirm the expression of the SNRPB2 protein in tumor tissues from clinical samples. The biological functions of SNRPB2 were assessed in vitro cell assay and in vivo tumor models. The molecular mechanisms were determined by correlation and gene set enrichment analysis. Western blot experiments validated involvement in signaling pathways. Results Our findings unveiled that SNRPB2 was upregulated at both mRNA and protein levels in ESCA, which was associated with the pathological progression of the disease. Additionally, SNRPB2 served as a robust prognostic biomarker, implicated in driving oncogenic functions in ESCA. It facilitated cell proliferation, migration, and invasion, transitioned the cell cycle, and inhibited apoptosis. Mechanistically, SNRPB2 activated genes associated with the β-catenin/c-Myc signaling pathway, such as β-catenin, c-Myc, CCNA2, CCNB1, CDK1, and CDK2. This activation also regulated the epithelial-to-mesenchymal transition (EMT), thereby facilitating the progression of ESCA. Conclusion Our findings demonstrate that SNRPB2 contributes to ESCA progression by regulating the β-catenin/c-Myc axis, suggesting its potential as a prognostic biomarker and therapeutic target for ESCA patients.
Collapse
Affiliation(s)
- Jiaqian Bao
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiong Tian
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yixiao Pan
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenyu Yang
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Meifu Gan
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Pathology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jingmin Zheng
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
8
|
Abid J, Al-Rawi MBA, Mahmood A, Li A, Jiang T. Identification and functional characterization of key biomarkers in diffuse large B-cell lymphoma: emphasis on STYX as a prognostic marker and therapeutic target. Hereditas 2025; 162:45. [PMID: 40128844 PMCID: PMC11931869 DOI: 10.1186/s41065-025-00411-w] [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: 11/23/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBC) is the most common subtype of non-Hodgkin lymphoma, characterized by its aggressive nature and poor prognosis in advanced stages. Despite advances in treatment, the molecular mechanisms driving DLBC progression remain incompletely understood, necessitating the identification of novel biomarkers for diagnosis and prognosis. In this study, we analyzed two publicly available datasets (GSE32018 and GSE56315) from the Gene Expression Omnibus database (GEO) to identify overlapping differentially expressed genes (DEGs). Later on, a comprehensive in silico and in vitro methodology was adopted to decipher the role of identify DEGs in DLBC. DEGs analysis of GSE32018 and GSE56315 datasets identified five overlapping gene: SP3, CSNK1A1, STYX, SIRT5, and MGEA5. Expression validation using the GEPIA2 database confirmed the upregulation of SP3, CSNK1A1, STYX, and SIRT5, and the downregulation of MGEA5 in DLBC tissues compared to normal controls. Furthermore, mutational analysis revealed that CSNK1A1 was the only gene among these DEGs to exhibit mutations, with a 2.7% mutation frequency in DLBC patients. Methylation analysis highlighted a negative correlation between DEGs methylation levels and mRNA expression, while survival analysis identified high STYX expression as significantly associated with poorer overall survival in DLBC patients. Functional assays demonstrated that STYX knockdown in U2932 cells led to reduced cell proliferation, colony formation, and enhanced wound healing, indicating STYX's pivotal role in DLBC cell survival and migration. Additionally, gene enrichment analysis revealed the involvement of these DEGs in key biological processes, including intracellular trafficking and myeloid progenitor cell differentiation. These findings emphasize the potential of SP3, CSNK1A1, STYX, SIRT5, and MGEA5 as biomarkers and therapeutic targets in DLBC, particularly highlighting STYX as a promising prognostic marker and potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Junaid Abid
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300222, China
| | - Mahmood Basil A Al-Rawi
- Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Ahmad Mahmood
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - An Li
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| | - Tiemin Jiang
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| |
Collapse
|
9
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
10
|
Saini N, Tiwari AK, Leahy R, Thorat N, Kulkarni A. Transforming brain cancer biomarker research with patinformatics and SWOT analysis. Drug Discov Today 2025; 30:104314. [PMID: 39971181 DOI: 10.1016/j.drudis.2025.104314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/29/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Brain cancer heterogeneity imposes significant challenges in diagnosis, causing high mortality. The lack of timely diagnosis intensifies these challenges, underscoring the need for improved diagnostics. Recent advancements in biomarker discovery have led to biomarker detection at ultra-low concentrations via multiplexing with biosensors, offering a promising avenue for the timely detection of brain cancer. Serving as a comprehensive resource, this review highlights the crucial role of primary biomarkers in brain cancer diagnosis via integration of patinformatics and SWOT analysis, thereby facilitating timely diagnosis and informed decision making. Furthermore, we aim to outline recent advances in brain cancer prognostics and management strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Neha Saini
- Symbiosis Centre for Nanoscience and Nanotechnology, Symbiosis International (Deemed University), Pune 412115, India
| | - Amit Kumar Tiwari
- Symbiosis Centre for Research and Innovation, Symbiosis International (Deemed University), Pune 412115, India; Patent Department R.K. Dewan and Co., Pune 411016 Maharashtra, India
| | - Robert Leahy
- Department of Physics and Bernal Institute University of Limerick, Castletroy, Limerick V94T9PX, Ireland
| | - Nanasaheb Thorat
- Department of Physics and Bernal Institute University of Limerick, Castletroy, Limerick V94T9PX, Ireland; Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick V94T9PX, Ireland.
| | - Atul Kulkarni
- Symbiosis Centre for Nanoscience and Nanotechnology, Symbiosis International (Deemed University), Pune 412115, India.
| |
Collapse
|
11
|
Bandara S, Raveendran S. Current Landscape and Future Directions in Cancer Immunotherapy: Therapies, Trials, and Challenges. Cancers (Basel) 2025; 17:821. [PMID: 40075668 PMCID: PMC11899461 DOI: 10.3390/cancers17050821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a leading global health challenge, placing immense burdens on individuals and healthcare systems. Despite advancements in traditional treatments, significant limitations persist, including treatment resistance, severe side effects, and disease recurrence. Immunotherapy has emerged as a promising alternative, leveraging the immune system to target and eliminate tumour cells. However, challenges such as immunotherapy resistance, patient response variability, and the need for improved biomarkers limit its widespread success. This review provides a comprehensive analysis of the current landscape of cancer immunotherapy, highlighting both FDA-approved therapies and novel approaches in clinical development. It explores immune checkpoint inhibitors, cell and gene therapies, monoclonal antibodies, and nanotechnology-driven strategies, offering insights into their mechanisms, efficacy, and limitations. By integrating emerging research and clinical advancements, this review underscores the need for continued innovation to optimise cancer immunotherapy and overcome existing treatment barriers.
Collapse
Affiliation(s)
- Shehani Bandara
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Sreejith Raveendran
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| |
Collapse
|
12
|
Attar M, Tash Shamsabadi F, Soltani A, Joghataei MT, Khandoozi SR, Teimourian S, Shahbazi M, Erfani-Moghadam V. MF59-based lipid nanocarriers for paclitaxel delivery: optimization and anticancer evaluation. Sci Rep 2025; 15:6583. [PMID: 39994380 PMCID: PMC11850822 DOI: 10.1038/s41598-025-91504-z] [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: 12/18/2024] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
Breast cancer is the most common invasive cancer in women worldwide, necessitating innovative therapeutic strategies to enhance treatment efficacy and safety. This study focuses on the development and optimization of novel paclitaxel (PTX)-loaded nanostructured lipid carriers (NLCs) that incorporate components of MF59, an oil-in-water emulsion adjuvant approved for use in influenza vaccines and known for its safety in humans. The formulation of these NLCs is designed to overcome significant challenges in PTX delivery, particularly its poor solubility and the side effects associated with traditional formulations containing Cremophor EL. We prepared two sets of NLC formulations using different liquid-to-solid lipid ratios through hot melt ultrasonication. Characterization of the selected formulations, NLCPre and NLCLec, was conducted using dynamic light scattering (DLS), scanning electron microscopy (SEM), Fourier-transform infrared (FT-IR) spectroscopy, and ultraviolet-visible (UV-Vis) spectroscopy. The mean diameters were 120.6 ± 36.4 nm and 112 ± 41.7 nm, with encapsulation efficiencies (EE) of 85% and 82%, and drug loading (DL) of 4.25% and 4.1%, respectively for NLCPre and NLCLec. In vitro cytotoxicity assays demonstrated that these MF59-based NLCs effectively target MCF-7 (Michigan Cancer Foundation) breast cancer cells while minimizing toxicity to normal HDF (human dermal fibroblasts) cells, thus enhancing the therapeutic index of PTX and offering promising clinical implications for breast cancer treatment.
Collapse
Affiliation(s)
- Marzieh Attar
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, 1416634793, Iran
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran
| | - Fatemeh Tash Shamsabadi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medical Sciences, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran
| | - Alireza Soltani
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, 1416634793, Iran
| | - Seyed Reza Khandoozi
- Cancer Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, Iran
| | - Shahram Teimourian
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, 1416634793, Iran.
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, 1416634793, Iran.
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran.
| | - Vahid Erfani-Moghadam
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran.
- Department of Medical Biotechnology, School of Advanced Technologies in Medical Sciences, Golestan University of Medical Sciences (GOUMS), Gorgan, 4934174515, Iran.
| |
Collapse
|
13
|
Huang H, Dong W, Qin X, Usama A, Cheema A, Deng C, Sarfaraz S, Pan Q, Alhomrani M, Alamri AS, ALSuhaymi N, Alghamdi SA, Alghamdi AA, Zheng S. Comprehensive pan-cancer analysis of LAMA3: implications for prognosis and immunotherapy. Am J Transl Res 2025; 17:1200-1222. [PMID: 40092111 PMCID: PMC11909552 DOI: 10.62347/qyjw2277] [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: 07/13/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES Laminin subunit alpha 3 (LAMA3) has been implicated in various cellular processes relevant to cancer progression, including cell proliferation, migration, and adhesion. In this study, we explored the expression, prognostic significance, and functional role of LAMA3 across multiple cancer types. METHODOLOGY The in silico analyses involve using various bioinformatics tools and databases, such as The Cancer Genome Atlas (TCGA), TIMER2.0, GEPIA2, UALCAN, Kaplan-Meier (KM) plotter, GENT2, Human Protein Atlas (HPA), OncoDB, Gene Set Cancer Analysis (GSCA), and TISIDB. The in vitro analyses include cell culture, gene knockdown, and assays for cell proliferation, colony formation, and wound healing. RESULTS Pan-cancer analysis revealed significant variations in LAMA3 expression, with upregulation observed in cancers such as pancreatic adenocarcinoma (PAAD) and stomach adenocarcinoma (STAD), and downregulation in breast cancer (BRCA) and colon adenocarcinoma (COAD). Prognostic analyses indicated high LAMA3 expression correlated with poor overall survival (OS) in PAAD and STAD, whereas low expression was associated with adverse outcomes in BRCA. Validation analysis confirmed differential expression and localized LAMA3 primarily to the endoplasmic reticulum. Analysis of clinical features in BRCA, PAAD, and STAD showed consistent expression trends across different stages, races, and age groups. Additionally, mutational and copy number variations (CNVs) analyses revealed prevalent heterozygous amplifications and deletions in LAMA3 across BRCA, PAAD, and STAD. Promoter methylation was inversely correlated with LAMA3 expression in BRCA, PAAD, and STAD, although survival outcomes were unaffected. Protein-protein interaction (PPI) and gene enrichment analyses indicated LAMA3's involvement in ECM-receptor interactions and PI3K-Akt signaling, pathways critical in cancer. Finally, functional assays following LAMA3 knockdown in HT-29 cells demonstrated reduced cell proliferation, colony formation, and wound healing, implicating LAMA3 in tumor growth and metastasis. CONCLUSION Overall, these findings suggest that LAMA3 plays a multifaceted role in tumorigenesis and holds potential as a prognostic biomarker and therapeutic target in multiple cancers.
Collapse
Affiliation(s)
- Hui Huang
- Department of Oncology, The Fifth Hospital of WuhanWuhan 430050, Hubei, China
| | - Wei Dong
- Department of Radiology, The Fifth Hospital of WuhanWuhan 430050, Hubei, China
| | - Xuan Qin
- Department of Radiology, The Fifth Hospital of WuhanWuhan 430050, Hubei, China
| | - Ali Usama
- Department of Internal Medicine, Wyckoff Heights Medical CenterBrooklyn, NY 11237, USA
| | - Anees Cheema
- Department of Internal Medicine, Wyckoff Heights Medical CenterBrooklyn, NY 11237, USA
| | - Chunlei Deng
- Department of Reproductive Medicine, Taihe Hospital Affiliated to Hubei University of MedicineShiyan 442000, Hubei, China
| | - Sara Sarfaraz
- Department of Bioinformatics, Faculty of Biomedical and Life Sciences, Kohsar UniversityMurree 47150, Pakistan
| | - Qingyun Pan
- Department of Endocrinology, The Fifth Hospital of WuhanWuhan 430050, Hubei, China
| | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura UniversityMakkah 21912, Saudi Arabia
| | - Saleh A Alghamdi
- Department of Clinical Laboratory Since, Medical Genetics, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Ahmad A Alghamdi
- Department of Clinical Laboratory Since, Medical Genetics, College of Applied Medical Sciences, Taif UniversityTaif 21944, Saudi Arabia
| | - Su Zheng
- Department of Rehabilitation, Taihe Hospital (Affiliated Hospital of Hubei University of Medical)Shiyan 442000, Hubei, China
| |
Collapse
|
14
|
Imran M, Altamimi ASA, Babu MA, Goyal K, Kaur I, Kumar S, Sharma N, Kumar MR, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H. Non-coding RNAs (ncRNAs) as therapeutic targets and biomarkers in oligodendroglioma. Pathol Res Pract 2024; 264:155708. [PMID: 39531874 DOI: 10.1016/j.prp.2024.155708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Oligodendrogliomas (ODGs) are neuroepithelial tumors that need personalized treatment plans because of their unique molecular and histological features. Non-coding RNAs form an epigenetic class of molecules that act as the first steps in gene regulation. They consist of microRNAs, long non-coding RNAs, and circular RNAs. These molecules significantly participate in ODG pathogenesis by regulating ODG initiation, progression, and treatment response. This review is designated to analyze the literature and describe the genomic profile of ODGs, the complex actions of ncRNAs in ODGs pathogenesis and treatment, and their roles as appropriate biomarkers and as one of the precision mechanisms action targets, such as antisense oligonucleotides, small interfering RNAs, gene therapy vectors, peptide nucleic acids, and small molecule inhibitors. Overall, ncRNAs considerably alter the pathological spectrum of ODGs by influencing fundamental processes in tumor biology. Applying ncRNAs in a clinical context exhibits promise for enhanced diagnosis and individualized therapeutic interventions. Nevertheless, the delivery efficacy and potential adverse "off-target" sequels retain the main obstacles undermining clinical potential. Continuous research and technological advancements in ncRNAs offer new insights and promising prospects for revolutionizing oligodendroglioma care, leading to better, personalized treatment outcomes.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India.
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Fadiyah Jadid Alanazi
- Center for Health Research, Northern Border University, Arar, Saudi Arabia; Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf City 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Division of Translational Health Research, Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|