101
|
S BR, Dhar R, Devi A. Exosomes-mediated CRISPR/Cas delivery: A cutting-edge frontier in cancer gene therapy. Gene 2025; 944:149296. [PMID: 39884405 DOI: 10.1016/j.gene.2025.149296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Cancer is considered the second most common disease globally. In the past few decades, many approaches have been proposed for cancer treatment. One among those is targeted therapy using CRISPR/Cas system which plays a significant role in translational research through gene editing. However, due to its inability to cope with specific targeting, off-target effects, and limited tumor penetration, it is very challenging to use this approach in cancer studies. To increase its efficacy, CRISPR components are engineered into the extracellular vesicles (EVs), especially exosomes (a subpopulation of EVs). Exosomes have a significant role in cellular communication. Exosomes-based CRISPR/Cas system transport for gene editing enhances specificity, reduces off-target effects, and improves the therapeutic potential. This review highlights the role of exosomes and the CRISPR/Cas system in cancer research, exosomes-based CRISPR delivery for cancer treatment, and its future orientation.
Collapse
Affiliation(s)
- Bhavanisha Rithiga S
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India.
| |
Collapse
|
102
|
Thomas NM, Alharbi M, Muripiti V, Banothu J. Quinoline and quinolone carboxamides: A review of anticancer activity with detailed structure-activity relationship analysis. Mol Divers 2025:10.1007/s11030-024-11092-4. [PMID: 39873887 DOI: 10.1007/s11030-024-11092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
Quinoline is a highly privileged scaffold with significant pharmacological potential. Introducing a carbonyl group into the quinoline ring generates a quinolone ring, which exhibits promising biological properties. Incorporating a carboxamide linkage at different positions within the quinoline and quinolone frameworks has proven an effective strategy for enhancing pharmacological properties, particularly anticancer potency. Consequently, various scientific communities have explored quinoline and quinolone carboxamides for their anticancer activities, introducing modifications at key positions. This review article aims to compile the anticancer activity of various quinoline and quinolone carboxamide derivatives, accompanied by a detailed structure-activity relationship (SAR) analysis. It also categorizes the data into activities of isolated/fused quinoline and quinolone carboxamide derivatives, which were further subclassified based on the mechanisms of anticancer action. Among the numerous derivatives studied, compounds 8, 19, 31, 34, 40, 68, 108, 116, and 132 have emerged as the most potent anticancer agents, making them strong candidates for further drug design and development. The mechanisms underlying the anticancer activity of these potent compounds have been identified as inhibitors of topoisomerase (8, 19, 31, and 34), protein kinase (40, 108, and 116), human dihydroorotate dehydrogenase (68), and as a cannabinoid receptor 2 agonist (132). We anticipate this review will be valuable to researchers engaged in the structural design and development of quinoline and quinolone carboxamide-based anticancer drugs with high efficacy.
Collapse
Affiliation(s)
- Neethu Mariam Thomas
- Department of Chemistry, National Institute of Technology Calicut, Kozhikode, 673601, Kerala, India
| | - Majed Alharbi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Venkanna Muripiti
- Department of Education, 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.
| |
Collapse
|
103
|
Budagova T, Efremova A, Usman N, Mokrousova D, Goldshtein D. Differentiating Induced Pluripotent Stem Cells into Natural Killer Cells for Adoptive Cell Immunotherapies-Comparative Characterization of Current Protocols. Int J Mol Sci 2025; 26:1107. [PMID: 39940874 PMCID: PMC11816922 DOI: 10.3390/ijms26031107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Cancers constitute a leading cause of mortality. Chimeric antigen receptor (CAR) cell therapies provide breakthrough solutions for various cancers while posing considerable risks of immunological side reactions. Of various cytotoxic lymphocyte subsets, natural killer (NK) cells are considered the least immunogenic. Obtaining viable NK cells with stable phenotypes in quantities sufficient for modification is technologically challenging. The candidate sources include primary mononuclear cell cultures and immortalized NK cell lines; alternatively, the clinical-grade NK cells can be differentiated from induced pluripotent stem cells (iPSCs) by a good manufacturing practice (GMP)-compatible xeno-free protocol. In this review, we analyze existing protocols for targeted differentiation of human iPSCs into NK cells with a focus on xeno-free requirements.
Collapse
Affiliation(s)
- Tatiana Budagova
- Research Centre for Medical Genetics, Moskvorechye Str. 1, Moscow 115522, Russia; (T.B.); (D.M.); (D.G.)
| | - Anna Efremova
- Research Centre for Medical Genetics, Moskvorechye Str. 1, Moscow 115522, Russia; (T.B.); (D.M.); (D.G.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, Moscow 117198, Russia
| | - Natalia Usman
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Samory Mashela Str. 1, Moscow 117198, Russia;
| | - Diana Mokrousova
- Research Centre for Medical Genetics, Moskvorechye Str. 1, Moscow 115522, Russia; (T.B.); (D.M.); (D.G.)
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, Moskvorechye Str. 1, Moscow 115522, Russia; (T.B.); (D.M.); (D.G.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, Moscow 117198, Russia
| |
Collapse
|
104
|
Fang Q, Chen S, Chen X, Zou W, Chen D, Huang Y, Wu C. Mature tertiary lymphoid structure associated CD103+ CD8+ Trm cells determined improved anti-tumor immune in breast cancer. Front Oncol 2025; 15:1480461. [PMID: 39926286 PMCID: PMC11802804 DOI: 10.3389/fonc.2025.1480461] [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: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Background Although tertiary lymphoid structures (TLS) play crucial roles in the anti-tumor immune response and are associated with favorable prognoses in many solid tumors, the precise mechanisms by which TLSs enhance anti-tumor immunity remain poorly understood. The current study aimed to explore the relationship between the maturity of tertiary lymphoid structures and their key immune cells in combating breast cancer. Patients and methods In this study, we utilized immunofluorescence and H&E staining to detect tumor-resident memory T cells (Trm) and assess the maturity of TLS, analyzing their distribution and proportion in an annotated cohort of 95 breast cancer patients. Results The presence of tumor-associated TLSs was correlated with an improved prognosis in patients with breast cancer. The proportion of CD8+CD103+ resident memory T cells and natural killer (NK) cells within the TLSs was significantly higher than that in areas outside of these structures. Additionally, the proportions of CD103+ CD8+ Trm cells and NK cells were significantly increased with the gradual maturation of TLS. Furthermore, the secretion function of effector molecules by CD8+ CD103+ Trm cells and NK cells within TLSs was significantly enhanced, indicating a strong correlation between the effector function of CD103+ CD8+ Trm and NK cells and the maturity of TLSs. Conclusion Our study identifies potential additional prognostic information for the clinical prognosis of breast cancer patients, underscoring the prognostic significance of immune cells within TLS, with a particular focus on CD103+ CD8+ Trm cells and NK cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chucheng Wu
- Huizhou Central People’s Hospital, Huizhou,
China
| |
Collapse
|
105
|
Gatti F, Perego G, Milano F, Calleri G, Giurioli B, Di Mattei VE. The Effects of Online Yoga Practice on Cancer Patients: A Systematic Review. Healthcare (Basel) 2025; 13:225. [PMID: 39942415 PMCID: PMC11817149 DOI: 10.3390/healthcare13030225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Cancer remains a leading cause of death, with 9.7 million deaths in 2022. Despite advancements in diagnosis and treatment, many cancer patients experience side effects that significantly impact their quality of life, including chronic pain, anxiety, depression, sleep disturbances, and cancer-related fatigue. Non-pharmacological interventions, such as yoga, have gained attention for their potential to reduce stress and improve overall well-being. However, barriers such as fatigue, pain, and transportation issues limit access to in-person yoga, leading to the growing adoption of online yoga as a viable alternative. Objective: This systematic review synthesizes research on the effectiveness of online yoga for cancer patients. A comprehensive search was conducted across Medline, PsycINFO, and Scopus databases on 24 October 2024. The methodological quality of the studies was assessed using the CASP Checklist. Of 6266 articles initially identified, 14 studies met the inclusion criteria, comprising qualitative (n = 4) and quantitative (n = 10) studies. Results: The results suggest that online yoga can improve stress and sleep quality, with moderate effects on anxiety, depression, and fatigue. However, variability in study designs and methodological limitations complicate the evaluation of its overall effectiveness. Conclusions: Online yoga offers a practical, accessible option for cancer patients unable to attend in-person sessions, showing the potential to enhance mental and physical health outcomes. Nevertheless, the variability in study methodologies highlights the need for more standardized research to establish its role as a supportive intervention in oncology care.
Collapse
Affiliation(s)
- Francesca Gatti
- School of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.G.); (G.P.); (V.E.D.M.)
| | - Gaia Perego
- School of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.G.); (G.P.); (V.E.D.M.)
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (B.G.)
| | - Francesca Milano
- Department of Psychology, University of Milano-Bicocca, 20132 Milan, Italy
| | - Gloria Calleri
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (B.G.)
| | - Bianca Giurioli
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (B.G.)
| | - Valentina Elisabetta Di Mattei
- School of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy; (F.G.); (G.P.); (V.E.D.M.)
- Clinical and Health Psychology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (B.G.)
| |
Collapse
|
106
|
Ramadan M, AlGhamdi S, Alsiary R. Analyzing the cancer mortality-to-incidence ratios and health expenditures in the aging population: a 20-year comparative study across high-income countries. FRONTIERS IN AGING 2025; 6:1506897. [PMID: 39917091 PMCID: PMC11794245 DOI: 10.3389/fragi.2025.1506897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025]
Abstract
Background The global burden of cancer is expected to increase by 60% over the next two decades, largely due to population aging. The study aims to examine the association between cancer mortality-to-incidence ratios (MIR) with healthcare expenditures (HE), and human development index score for individuals 70 years old or older. Method This is an epidemiological study using publicly available data from the Global Burden of Disease (GBD) for six over the years 1990-2019. A generalized linear model was employed to examine the association between MIR, and health expenditures and health development index score. Results Included countries showed a statistically significant negative association between MIR and both HE, and HE, indicating that higher HDI and HE are associated with decreased MIR with the highest decrease was for China, the coefficient for HDI is -1.29 (95% CI: -1.35 to -1.24, p < 0.0001), the coefficient for HE is -0.103 (95% CI: -0.17 to -0.03, p < 0.0001). There are variations exist in MIRs between high and low health expenditure countries for each cancer type. Conclusion The study reveals a significant impact of HE and HDI on cancer outcomes in older adults. Variations between high and low HE nations highlight potentially improved cancer outcomes in high HE countries. Considering the anticipated growth in the aging population worldwide, a rise in cancer cases is expected among older individuals. The implications are profound, suggesting an impending strain on healthcare systems, particularly in nations with a high proportion of elderly and low health expenditures.
Collapse
Affiliation(s)
- Majed Ramadan
- Population Health Research Section, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard–Health Affairs, Jeddah, Saudi Arabia
| | - Shadell AlGhamdi
- College of medicine, King Saud bin Abdulaziz University for Health Science, Ministry of National Guard–Health Affairs, Jeddah, Saudi Arabia
| | - Rawiah Alsiary
- Department of Cellular Therapy and Cancer Research, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard–Health Affairs, Jeddah, Saudi Arabia
| |
Collapse
|
107
|
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
|
108
|
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
|
109
|
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
|
110
|
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
|
111
|
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
|
112
|
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
|
113
|
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
|
114
|
Hien LT, Hieu PT, Toan DN. An Efficient 3D Convolutional Neural Network for Dose Prediction in Cancer Radiotherapy from CT Images. Diagnostics (Basel) 2025; 15:177. [PMID: 39857061 PMCID: PMC11765056 DOI: 10.3390/diagnostics15020177] [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: 11/18/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: Cancer is a highly lethal disease with a significantly high mortality rate. One of the most commonly used methods for treatment is radiation therapy. However, cancer treatment using radiotherapy is a time-consuming process that requires significant manual work from planners and doctors. In radiation therapy treatment planning, determining the dose distribution for each of the regions of the patient's body is one of the most difficult and important tasks. Nowadays, artificial intelligence has shown promising results in improving the quality of disease treatment, particularly in cancer radiation therapy. Objectives: The main objective of this study is to build a high-performance deep learning model for predicting radiation therapy doses for cancer and to develop software to easily manipulate and use this model. Materials and Methods: In this paper, we propose a custom 3D convolutional neural network model with a U-Net-based architecture to automatically predict radiation doses during cancer radiation therapy from CT images. To ensure that the predicted doses do not have negative values, which are not valid for radiation doses, a rectified linear unit (ReLU) function is applied to the output to convert negative values to zero. Additionally, a proposed loss function based on a dose-volume histogram is used to train the model, ensuring that the predicted dose concentrations are highly meaningful in terms of radiation therapy. The model is developed using the OpenKBP challenge dataset, which consists of 200, 100, and 40 head and neck cancer patients for training, testing, and validation, respectively. Before the training phase, preprocessing and augmentation techniques, such as standardization, translation, and flipping, are applied to the training set. During the training phase, a cosine annealing scheduler is applied to update the learning rate. Results and Conclusions: Our model achieved strong performance, with a good DVH score (1.444 Gy) on the test dataset, compared to previous studies and state-of-the-art models. In addition, we developed software to display the dose maps predicted by the proposed model for each 2D slice in order to facilitate usage and observation. These results may help doctors in treating cancer with radiation therapy in terms of both time and effectiveness.
Collapse
Affiliation(s)
- Lam Thanh Hien
- Faculty of Information Technology, Lac Hong University, Huynh Van Nghe, Bien Hoa 76120, Vietnam;
| | - Pham Trung Hieu
- Institute of Information Technology, Vietnam Academy of Science and Technology, Hoang Quoc Viet, Hanoi 10072, Vietnam;
| | - Do Nang Toan
- Institute of Information Technology, Vietnam Academy of Science and Technology, Hoang Quoc Viet, Hanoi 10072, Vietnam;
| |
Collapse
|
115
|
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
|
116
|
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
|
117
|
Munyayi TA, Crous A. Advancing Cancer Drug Delivery with Nanoparticles: Challenges and Prospects in Mathematical Modeling for In Vivo and In Vitro Systems. Cancers (Basel) 2025; 17:198. [PMID: 39857980 PMCID: PMC11763932 DOI: 10.3390/cancers17020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Mathematical models are crucial for predicting the behavior of drug conjugate nanoparticles and optimizing drug delivery systems in cancer therapy. These models simulate interactions among nanoparticle properties, tumor characteristics, and physiological conditions, including drug resistance and targeting specificity. However, they often rely on assumptions that may not accurately reflect in vivo conditions. In vitro studies, while useful, may not fully capture the complexities of the in vivo environment, leading to an overestimation of nanoparticle-based therapy effectiveness. Advancements in mathematical modeling, supported by preclinical data and artificial intelligence, are vital for refining nanoparticle-based therapies and improving their translation into effective clinical treatments.
Collapse
Affiliation(s)
| | - Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| |
Collapse
|
118
|
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
|
119
|
Tang S, McGinnis R, Cao Z, Baker, Jr. JR, Xu Z, Wang S. Ultrasound-Guided Histotripsy Triggers the Release of Tumor-Associated Antigens from Breast Cancers. Cancers (Basel) 2025; 17:183. [PMID: 39857965 PMCID: PMC11764245 DOI: 10.3390/cancers17020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: There is increasing evidence to indicate that histotripsy treatment can enhance the host anti-tumor immune responses both locally at the targeting tumor site as well as systemically from abscopal effects. Histotripsy is a non-invasive ultrasound ablation technology that mechanically disrupts target tissue via cavitation. A key factor contributing to histotripsy-induced abscopal effects is believed to be the release of tumor-specific antigens (TSAs) or tumor-associated antigens (TAAs) that induce a systemic immune response. In this study, we studied the effect of histotripsy treatment on the release of HER2, a well-defined TAA target for cancer immunotherapy. Methods: A range of doses of histotripsy administered to HER2-postive mammary tumor cells in an in vitro cell culture system and an ex vivo tumor were applied. In addition, a single dose of histotripsy was used for an in vivo murine tumor model. The released proteins, and specifically HER2, in both tumor cell-free supernatants and tumor cell pellets were analyzed by a BCA protein assay, an ultra-performance liquid chromatography (UPLC) assay, and Western blot. Results: Our results showed that histotripsy could significantly trigger the release of HER2 proteins in the current study. The level of HER2 proteins was actually higher in tumor cell-free supernatants than in tumor cell pellets, suggesting that HER2 was released from the intracellular domain into the extracellular compartment. Furthermore, proportionally more HER2 protein was released at higher histotripsy doses, indicating free HER2 was histotripsy-dose-dependent. Conclusions: In conclusion, we have qualitatively and quantitatively demonstrated that histotripsy treatment triggers the release of HER2 from the tumor cells into the extracellular compartment. The histotripsy-mediated release of HER2 antigens provides important insights into the mechanism underlying its immunostimulation and suggests the potential of TSA/TAA-based immunotherapies in numerous cancer types.
Collapse
Affiliation(s)
- Shengzhuang Tang
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - Reliza McGinnis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.M.); (Z.X.)
| | - Zhengyi Cao
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - James R. Baker, Jr.
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.M.); (Z.X.)
| | - Suhe Wang
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| |
Collapse
|
120
|
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
|
121
|
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
|
122
|
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
|
123
|
Thirumalai A, Girigoswami K, Harini K, Kiran V, Durgadevi P, Girigoswami A. Natural polymer derivative-based pH-responsive nanoformulations with entrapped diketo-tautomers of 5-fluorouracil for enhanced cancer therapy. ADMET AND DMPK 2025; 13:2554. [PMID: 40161888 PMCID: PMC11954142 DOI: 10.5599/admet.2554] [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: 11/01/2024] [Revised: 12/27/2024] [Indexed: 04/02/2025] Open
Abstract
Background and purpose Despite significant advancements in cancer therapies, chemotherapeutics continue to be the mainstay for treating cancer patients, with 5-fluorouracil (5-FU) being commonly used for various cancers. However, its limited ability to penetrate cell membranes and its short half-life, caused by rapid metabolism, necessitate frequent administration of high doses to maintain effective therapeutic levels. This study aimed to synthesize oxidized sodium alginate (OSA) derivatives to create OSA nanoparticles loaded with 5-FU (OSANP@ 5-FU), promoting diketo tautomers, and evaluate their photophysical properties, release profile, and anticancer activity with minimal toxicity. Experimental approach The investigation encompassed physicochemical characterization, encapsulation efficiency, 5-FU release kinetics at pH 2.2 and 7.4, cell viability assessment via MTT assay in V79 cells, and in vitro anticancer efficacy in the A375 cell line. Key results Steady-state absorption and emission confirmed the presence of advantageous diketone tautomers of 5-FU, indicating radiative transitions from the second singlet excited state to the ground state (S2→S0) and the drug's encapsulation within the polymeric nanostructure. Dynamic light scattering revealed that OSA nanoparticles, initially 177.8 nm, grew to 226.6 nm after encapsulating 5-FU, retaining high zeta potential for stability. With a 68% encapsulation efficiency, in vitro studies showed 46 to 54 % of 5-FU released across different pH levels within 510 minutes. Conclusion In acidic conditions, there is a greater release of 5-FU than neutral pH levels, indicating a pH-responsive release profile beneficial for cancer treatment, with the release mechanism of OSANPs following Fickian diffusion as identified by a Korsmeyer-Peppas mathematical model and the formulation showing improved therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN-603103, India
| |
Collapse
|
124
|
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
|
125
|
Sabokrouh A, Ghaffari N, Karimi Tafreshi Z, Atabi F, Sharifi Z, Shokouhi Mostavafi SK. Comparison of Anticancer Effects between Platinum Levetiracetam and Platinum Azidothymidine through the Expression of Biomarker Genes on Cancer Cell Lines. Avicenna J Med Biotechnol 2025; 17:24-30. [PMID: 40094094 PMCID: PMC11910025 DOI: 10.18502/ajmb.v17i1.17674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/02/2024] [Indexed: 03/19/2025] Open
Abstract
Background The utilization of biomarkers is a way to assess the efficacy of recently created anticancer drugs. MiRNAs, telomerase, and Bcl-2 are extensively utilized as biomarkers for this purpose. This study aims to evaluate the comparison of the newly synthesized platinum compounds such as Platinum Azidothymidine (Pt-AZT) with Platinum Levetiracetam (Pt-Lev) on HepG2 cancer cell lines via the biomarkers. Methods In this study, cells were divided into four groups: Group A (HDF cells) were the normal negative control group, group B were HepG2 untreated cancer cells, and groups C and D were treated cancer cells with Pt-AZT and Pt-Lev, respectively. After evaluating the LC50 for the drugs by MTT test, the relative gene expression of the biomarkers was determined by qPCR. Results The results showed a significant decrease for antiapoptotic genes including miRNA-21 (5.1±0.014), telomerase (0.56±0.48), Bcl-2 (0.41±0.276) in group D, whereas in group C was more than group D for miRNA-21 (6.0±0.141), telomerase (3.49±0.231), Bcl-2 (4.93±0.276) also there was a significant increase in miRNA-122 (33.97±0.04) in group D, whereas in group C was (28.36±0.007) and so lower than group D. Most of the investigated groups showed a significant difference (p<0.05). In addition, there were widespread apoptotic regions in Pt-Lev treatment compared to Pt-AZT. Conclusion The advantages of using Pt-Lev were more powerful anticancer effects on biomarkers through inhibition of antiapoptotic and stimulation proapoptotic factors and also lower side effects and lower drug resistance than Pt-AZT; therefore, it can be considered a more effective anti-cancer therapy.
Collapse
Affiliation(s)
- Abdolreza Sabokrouh
- Department of Biochemistry, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nasim Ghaffari
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Karimi Tafreshi
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Freshteh Atabi
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zohreh Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | | |
Collapse
|
126
|
Gencsoy Eker S, Inetas Yengin G, Tatar C, Oktem G. A Comprehensive Review of the Mechanisms and Clinical Development of Monoclonal Antibodies in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:181-203. [PMID: 39666264 DOI: 10.1007/5584_2024_838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Cancer is still the disease that ranks first in human mortality in the twenty-first century. In the last 20 years, the concept of molecular targeted therapy has come to the fore with the use of small molecule agents or signal transduction inhibitors that show anticancer effects for certain types of cancer. Monoclonal antibodies, which have a therapeutic effect, especially by providing signal transduction inhibition, are used clinically as first-line treatment in various types of cancer. Molecular targeted therapies are critical for eliminating the adverse effects and drug resistance problems that occur in traditional cancer treatments. This review summarizes current information on various targeted therapeutic agents, including the structure and classification of monoclonal antibodies, their production methods and mechanisms of action, the monoclonal antibodies used in clinical trials, the complement system mechanism and cancer relationship, and the relationship between complement-dependent cytotoxicity and monoclonal antibodies.
Collapse
Affiliation(s)
- Selen Gencsoy Eker
- Department of Stem Cell, Graduate School of Health Sciences, Ege University, Izmir, Turkey
| | - Gizem Inetas Yengin
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Cansu Tatar
- Department of Molecular Biology and Genetics, Yildiz Technical University, Istanbul, Turkey
| | - Gulperi Oktem
- Department of Stem Cell, Graduate School of Health Sciences, Ege University, Izmir, Turkey.
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir, Turkey.
| |
Collapse
|
127
|
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
|
128
|
Bandaru PK, Nidasanametla SKR, Shyamala P. Amide Functionalized Novel Pyrrolo-pyrimidine Derivative as Anticancer Agents: Synthesis, Characterization and Molecular Docking Studies. Anticancer Agents Med Chem 2025; 25:420-432. [PMID: 39506418 DOI: 10.2174/0118715206333935241004070350] [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: 06/23/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND The development of new therapies targeting crucial kinases involved in cancer progression is a promising area of research. Pyrazolo pyrimidine derivatives have emerged as potential candidates for this purpose. OBJECTIVE This study aims to synthesize pyrazolo pyrimidine derivatives (5a-5r), evaluate their molecular docking against key kinases, and assess their anticancer activity. METHODS The synthesis involved a multi-step procedure starting with the cyclization of 6-amino-2- methylpyrimidin-4(3H)-one (1) to form 2-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-4-ol (2). This was followed by chlorination to yield 4-chloro-2-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidine (3) and nucleophilic substitution to produce 2-methyl-4,7-dihydro-3H-pyrrolo[2,3-d]pyrimidin-4-amine (4). The final derivatives (5a-5r) were synthesized through amide bond formation with various carboxylic acids using DCC and DMAP. Structural elucidation was confirmed via NMR, mass spectrometry, and HRMS. Molecular docking studies were conducted against Janus kinase 1 (JAK1), Janus kinase 2 (JAK2), and cyclin-dependent kinase 4 (CDK4). Anticancer activity was evaluated against MCF-7, SET-2, and HCT-116 cell lines. RESULTS Structural elucidation confirmed the successful synthesis of the derivatives. Molecular docking studies revealed promising binding affinities for selected derivatives, particularly those with heterocyclic substitutions. Anticancer activity evaluation showed diverse potency profiles, with several derivatives demonstrating IC50 values comparable to the reference drug, doxorubicin. Derivatives featuring nitro and heterocyclic moieties exhibited significant anticancer activity. CONCLUSION The synthesized pyrazolo pyrimidine derivatives showed potential as lead compounds for further development due to their promising binding affinities and significant anticancer activity, particularly those with nitro and heterocyclic moieties.
Collapse
Affiliation(s)
| | | | - Pulipaka Shyamala
- Department of Chemistry, Andhra University, Vishakhapatnam, Andhra Pradesh, India
| |
Collapse
|
129
|
Sekeroglu ZA, Sekeroglu V. A Review on Patient-derived 3D Micro Cancer Approach for Drug Screen in Personalized Cancer Medicine. Curr Cancer Drug Targets 2025; 25:118-130. [PMID: 38445692 DOI: 10.2174/0115680096285910240206044830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 03/07/2024]
Abstract
Precision medicine in oncology aims to identify an individualized treatment plan based on genomic alterations in a patient's tumor. It helps to select the most beneficial therapy for an individual patient. As it is now known that no patient's cancer is the same, and therefore, different patients may respond differently to conventional treatments, precision medicine, which replaces the one-size-fits-all approach, supports the development of tailored treatments for specific cancers of different patients. Patient-specific organoid or spheroid models as 3D cell culture models are very promising for predicting resistance to anti-cancer drugs and for identifying the most effective cancer therapy for high-throughput drug screening combined with genomic analysis in personalized medicine. Because tumor spheroids incorporate many features of solid tumors and reflect resistance to drugs and radiation, as in human cancers, they are widely used in drug screening studies. Testing patient-derived 3D cancer spheroids with some anticancer drugs based on information from molecular profiling can reveal the sensitivity of tumor cells to drugs and provide the right compounds to be effective against resistant cells. Given that many patients do not respond to standard treatments, patient-specific treatments will be more effective, less toxic. They will affect survival better compared to the standard approach used for all patients.
Collapse
Affiliation(s)
- Zulal Atlı Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Sekeroglu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
130
|
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
|
131
|
Allen D, Szoo MJ, van Bergen TD, Seppelin A, Oh J, Saad MA. Near-infrared photoimmunotherapy: mechanisms, applications, and future perspectives in cancer research. Antib Ther 2025; 8:68-85. [PMID: 39958565 PMCID: PMC11826922 DOI: 10.1093/abt/tbaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/01/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
Photoimmunotherapy (PIT) involves the targeted delivery of a photosensitizer through antibody conjugation, which, upon binding to its cellular target and activation by external irradiation, induces localized toxicity. This approach addresses several limitations of conventional cancer therapies, such as chemo- and radiotherapies, which result in off-target effects that significantly reduce patient quality of life. Furthermore, PIT improves on the challenges encountered with photodynamic therapy (PDT), such as nonspecific localization of the photosensitizer, which often results in unintended toxicities. Although PIT was first proposed in the early 1980s, its clinical applications have been constrained by limitations in antibody engineering, conjugation chemistries, and optical technologies. However, recent advances in antibody-drug conjugate (ADC) research and the emergence of sophisticated laser technologies have greatly benefited the broader applicability of PIT. Notably, the first near-infrared photoimmunotherapy (NIR-PIT) treatment for head and neck cancer has been approved in Japan and is currently in phase III clinical trials in the USA. A significant advantage of PIT over traditional ADCs in cancer management is the agnostic nature of PDT, making it more adaptable to different tumor types. Specifically, PIT can act on cancer stem cells and cancer cells displaying treatment resistance and aggressive phenotypes-a capability beyond the scope of ADCs alone. This review provides an overview of the mechanism of action of NIR-PIT, highlighting its adaptability and application in cancer therapeutics, and concludes by exploring the potential of PIT in advancing cancer treatments.
Collapse
Affiliation(s)
- Derek Allen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Madeline JoAnna Szoo
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States
- Department of Biochemistry, Northeastern University, Boston, MA 02115, United States
| | - Tessa D van Bergen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Ani Seppelin
- Department of Biochemistry, Northeastern University, Boston, MA 02115, United States
| | - Jeonghyun Oh
- Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
132
|
Mukherjee D, Roy D, Thakur S. Transforming Cancer Care: The Impact of AI-driven Strategies. Curr Cancer Drug Targets 2025; 25:204-207. [PMID: 38988158 DOI: 10.2174/0115680096323564240703102748] [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: 05/01/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024]
Abstract
AI is a critical component in healthcare, especially in the application of precision medicine where patients' characteristics, including genetic makeup, determine the treatment options that should be implemented. AI sorts big data, predicting people's reactions to specific treatments, the right combinations of drugs, and possible side effects, therefore increasing the efficiency of the treatment process and decreasing negative outcomes. This article briefly presents the ethical issues and concerns that might arise due to the integration of AI in society, such as the privacy of data, the issues of bias in the algorithms, and the issues of interpretability of the AI systems. Nevertheless, there is no doubt that AI can bring qualitative changes in cancer care based on its potential to enhance patient prognosis and reduce health care costs, as well as become a defining feature of the standard of care.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Debajyoti Roy
- Department of Pharmacognosy, C.V. Raman Global University, Bhubaneswar, 752054, Odisha, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
133
|
Soniya N, Soumya V, Shivlingrao MD, M M, Meeramol C. Therapeutic Potental of Quinolin-2 H-one Hybrids as Anticancer Agents. Mini Rev Med Chem 2025; 25:386-402. [PMID: 39323349 DOI: 10.2174/0113895575305597240912192037] [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: 02/24/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/27/2024]
Abstract
The statistical data related to cancer in recent years has shown a great increase in the number of cases and is likely to further increase in the future. Even after seeking thorough knowledge on the aetiology of cancer and related disorders and attempting to cure it by various methods like gene therapy, T cell therapy, chemotherapy, surgery, hormone therapy, and photodynamic therapy, there has always been disappointment concerning the survival rate. Hence, there is still a great urge for the discovery of novel drugs for the treatment of cancer. Chemotherapy being one of the widely used methods, several drug entities possessing anticancer properties are already in the market but none of them is known to show good efficacy which necessitates researchers to design newer drugs for the treatment of cancer. The urge to synthesize novel anticancer entities directed researchers towards molecular hybridization as one of the novel methods for designing newer drugs. Literature reveals wide research carried out on quinolin-2-one hybrids, possessing anticancer properties through different mechanisms. Tipifarnib and Dovitinib are quinolin-2-one hybrids used to treat cancer, possessing imidazole and benzimidazole heterocyclic rings. Different heterocyclic scaffolds such as pyrone, pyrrole, pyrimidine, pyridine, thiazole, and pyrazole in combination with heterocyclic quinolin-2-one have shown high potential to become lead for newer anticancer agents with better and wider therapeutic properties and lesser side effects. The current review presents information on the different quinolin-2-one hybrids and their effect on different cancer cell lines. It also imparts knowledge of the structural requirements for designing novel anticancer agents.
Collapse
Affiliation(s)
- Naik Soniya
- Department of Pharmaceutical Chemistry, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
- Department of Pharmaceutical Chemistry, PESs Rajaram and Tarabai Bandekar College of Pharmacy, Farmagudi, Ponda, Goa, 403 401, India
| | - Vasu Soumya
- Department of Pharmaceutical Chemistry, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Mamle Desai Shivlingrao
- Department of Pharmaceutical Chemistry, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Manickavasagam M
- Department of Oncology, SRMC & RI, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| | - Chellappan Meeramol
- Department of Pharmaceutical Chemistry, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, 600116, India
| |
Collapse
|
134
|
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 LettersIstanbul Technical UniversityIstanbulTurkiye
- Department of Molecular Biology and Genetics, Graduate School of Science and EngineeringYildiz Technical UniversityIstanbulTurkiye
| | - Fatma Sayan Poyraz
- Department of Molecular Biology and Genetics, Faculty of Arts and SciencesYildiz Technical UniversityIstanbulTurkiye
| | - Seda Susgun
- Department of Genetics, Institute of Health SciencesIstanbul UniversityIstanbulTurkiye
- Department of Medical Biology, Faculty of MedicineBezmialem Vakif UniversityIstanbulTurkiye
| | - Emrah Yucesan
- Department of Neurogenetics, Institute of Neurological SciencesIstanbul University‐CerrahpasaIstanbulTurkiye
| | - Banu Mansuroglu
- Department of Molecular Biology and Genetics, Faculty of Arts and SciencesYildiz Technical UniversityIstanbulTurkiye
| |
Collapse
|
135
|
Singer M, Hamdy R, Elsayed TM, Husseiny MI. The Mechanisms and Therapeutic Implications of Metabolic Communication in the Tumor-Immune Microenvironment. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:291-315. [DOI: 10.1007/978-981-96-1305-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
136
|
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
|
137
|
Heriz MH, Razzak Mahmood AA, Tahtamouni LH, Al-Sakhen MF, Kanaan SI, Saleh KM, Yasin SR. New Carbothioamide and Carboxamide Derivatives of 3-Phenoxybenzoic Acid as Potent VEGFR-2 Inhibitors: Synthesis, Molecular Docking, and Cytotoxicity Assessment. Curr Cancer Drug Targets 2025; 25:412-430. [PMID: 38747227 DOI: 10.2174/0115680096307334240429050730] [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: 01/30/2024] [Revised: 03/16/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2025]
Abstract
INTRODUCTION/BACKGROUND Because of the well-established link between angiogenesis and tumor development, the use of antiangiogenic therapeutics, such as those targeting VEGFR-2, presents a promising approach to cancer treatment. In the current study, a set of five hydrazine-1- carbothioamide (compounds 3a-e) and three hydrazine-1-carboxamide derivatives (compounds 4a-c) were successfully synthesized from 3-phenoxybenzoic acid. These compounds were specially created as antiproliferative agents with the goal of targeting cancer cells by inhibiting VEGFR- 2 tyrosine kinase. MATERIALS AND METHODS The new derivatives were synthesized by conventional organic methods, and their structure was versified by IR, 1HNMR, 13CNMR, and mass spectroscopy. In silico investigation was carried out to identify the compounds' target, molecular similarity, ADMET, and toxicity profile. The cytotoxic activity of the prepared compounds was evaluated in vitro against three human cancer cell lines (DLD1 colorectal adenocarcinoma, HeLa cervical cancer, and HepG2 hepatocellular carcinoma). The effects of the leading compound on cell cycle progression and apoptosis induction were investigated by flow cytometry, and the specific apoptotic pathway triggered by the treatment was evaluated by RT-PCR and immunoblotting. Finally, the inhibitory activities of the new compounds against VEGFR-2 was measured. RESULTS The designed derivatives exhibited comparable binding positions and interactions to the VEGFR-2 binding site to that of sorafenib (a standard VEGFR-2 tyrosine kinase inhibitor), as determined by molecular docking analysis. Compound 4b was the most cytotoxic compound, achieving the lowest IC50 against HeLa cells. Compound 4b, a strong representative of the synthesized series, induced cell cycle arrest at the G2/M phase, increased the proportion of necrotic and apoptotic HeLa cells, and activated caspase 3. The EC50 value of compound 4b against VEGFR-2 kinase activity was comparable to sorafenib's. CONCLUSION Overall, the findings suggest that compound 4b has a promising future as a starting point for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Mohammad Hamza Heriz
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Al-Zahraa University for Women, Karbala-Baghdad Street, Karbala, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, Bab-Almoudam, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Mai F Al-Sakhen
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Sana I Kanaan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Khaled M Saleh
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Salem R Yasin
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
138
|
Naseem N, Kushwaha P, Haider F. Leveraging nanostructured lipid carriers to enhance targeted delivery and efficacy in breast cancer therapy: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:449-468. [PMID: 39196394 DOI: 10.1007/s00210-024-03408-w] [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: 05/29/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
Cancer, characterized by uncontrolled cell growth and proliferation, continues to be a major global health concern. Breast cancer, the most commonly diagnosed cancer among women, remains a leading cause of cancer-related deaths worldwide. Conventional treatment modalities such as surgery, radiation, and chemotherapy have made significant strides in improving patient outcomes. However, these approaches often face challenges such as limited efficacy, systemic toxicity, and multidrug resistance. Nanotechnology has emerged as a promising avenue for revolutionizing cancer therapy, offering targeted drug delivery, enhanced efficacy, and reduced side effects. Among the various nanocarrier systems, nanostructured lipid carriers (NLCs) have gained considerable attention for their unique advantages. Comprising a blend of solid and liquid lipids, NLCs offer improved drug loading capacity, enhanced stability, sustained release, and biocompatibility. This manuscript provides a comprehensive overview of the role of NLCs in breast cancer management, covering their formulation, methods of preparation, advantages, and disadvantages. Additionally, several studies are presented to illustrate the efficacy of NLCs in delivering anticancer drugs to breast tumors. These studies demonstrate the ability of NLCs to enhance drug cytotoxicity, improve tumor suppression, and minimize systemic toxicity. This manuscript aims to contribute to the existing literature by consolidating current knowledge and providing insights into the future directions of NLC-based therapeutics in breast cancer management.
Collapse
Affiliation(s)
- Nazish Naseem
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| | - Poonam Kushwaha
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India.
| | - Faheem Haider
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| |
Collapse
|
139
|
Amrutkar RD, Amesar MV, Chavan LB, Baviskar NS, Bhamare VG. Precision Targeting of BET Proteins - Navigating Disease Pathways, Inhibitor Insights, and Shaping Therapeutic Frontiers: A Comprehensive Review. Curr Drug Targets 2025; 26:147-166. [PMID: 39385413 DOI: 10.2174/0113894501304747240823111337] [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: 03/28/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 10/12/2024]
Abstract
The family of proteins known as Bromodomain and Extra-Terminal (BET) proteins has become a key participant in the control of gene expression, having a significant impact on numerous physiological and pathological mechanisms. This review offers a thorough investigation of the BET protein family, clarifying its various roles in essential cellular processes and its connection to a variety of illnesses, from inflammatory disorders to cancer. The article explores the structural and functional features of BET proteins, emphasizing their special bromodomain modules that control chromatin dynamics by identifying acetylated histones. BET proteins' complex roles in the development of cardiovascular, neurodegenerative, and cancer diseases are carefully investigated, providing insight into possible treatment avenues. In addition, the review carefully examines the history and relevance of BET inhibitors, demonstrating their capacity to modify gene expression profiles and specifically target BET proteins. The encouraging outcomes of preclinical and clinical research highlight BET inhibitors' therapeutic potential across a range of disease contexts. The article summarizes the state of BET inhibitors today and makes predictions about the challenges and future directions of the field. This article provides insights into the changing field of BET protein-targeted interventions by discussing the potential of personalized medicine and combination therapies involving BET inhibitors. This thorough analysis combines many aspects of BET proteins, such as their physiological roles and their roles in pathophysiological conditions. As such, it is an invaluable tool for scientists and medical professionals who are trying to figure out how to treat patients by using this fascinating protein family.
Collapse
Affiliation(s)
- Rakesh D Amrutkar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Mehul V Amesar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Lokesh B Chavan
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Nilesh S Baviskar
- Department of Pharmaceutical Chemistry, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| | - Vaibhav G Bhamare
- Department of Pharmaceutics, K. K. Wagh College of Pharmacy, Panchavati Nasik, India
| |
Collapse
|
140
|
Zuo Y, Li P, Wang W, Xu C, Xu S, Sung HHY, Sun J, Jin G, Wang W, Kwok RTK, Lam JWY, Tang BZ. Tumor Site-Specific In Vivo Theranostics Enabled by Microenvironment-Dependent Chemical Transformation and Self-Amplifying Effect. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409506. [PMID: 39612249 PMCID: PMC11789590 DOI: 10.1002/advs.202409506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/12/2024] [Indexed: 12/01/2024]
Abstract
Precise tumor diagnosis and treatment remain complex challenges. While numerous fluorescent probes have been developed for tumor-specific imaging and therapy, few exhibit effective function in vivo. Herein, a probe called TQ-H2 is designed that can realize robust theranostic effects both in vitro and in vivo. In vitro, TQ-H2 specifically targets the lysosome and reacts with hydroxyl radical (·OH) to generate TQ-HA, which lights up the cells. TQ-HA generates reactive oxygen species (ROS) under light irradiation, enabling the simultaneous induction and monitoring of apoptosis and ferroptosis in tumor cells. Remarkably, TQ-HA also acts as a self-amplifier, autocatalytically activating TQ-H2 by generating ·OH under light exposure. This self-amplification aligns with the tumor microenvironment, where TQ-H2 undergoes chemical transformation, distinguishing tumors from healthy tissue via near-infrared (NIR) fluorescence imaging. Furthermore, ROS generated by TQ-HA effectively kills tumor cells and inhibits tumor growth without harming normal cells. This study offers a promising strategy for targeted tumor theranostics using self-amplifying microenvironment-responsive fluorescent probes.
Collapse
Affiliation(s)
- Yunfei Zuo
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
| | - Pei Li
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
- National Clinical Research Center for Infectious DiseasesShenzhen Third People's HospitalSouthern University of Science and TechnologyShenzhenGuangdong518112China
| | - Wen‐Jin Wang
- China Clinical Translational Research Center of Aggregation‐Induced EmissionThe Second Affiliated HospitalSchool of MedicineSchool of Science and EngineeringShenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong KongShenzhen (CUHK‐Shenzhen)Guangdong518172China
| | - Changhuo Xu
- MOE Frontiers Science Center for Precision OncologyFaculty of Health SciencesUniversity of MacauMacao999078China
| | - Shuting Xu
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
| | - Herman H. Y. Sung
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
| | - Jianwei Sun
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049China
| | - Weiping Wang
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
| | - Ryan T. K. Kwok
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
| | - Jacky W. Y. Lam
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
| | - Ben Zhong Tang
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular Neuroscienceand Department of Chemical and Biological EngineeringThe Hong Kong University of Science & TechnologyClear Water BayKowloonHong Kong999077P. R. China
- China Clinical Translational Research Center of Aggregation‐Induced EmissionThe Second Affiliated HospitalSchool of MedicineSchool of Science and EngineeringShenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong KongShenzhen (CUHK‐Shenzhen)Guangdong518172China
| |
Collapse
|
141
|
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
|
142
|
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
|
143
|
Sahu C, Sahu RK, Roy A. A Review on Nanotechnologically Derived Phytomedicines for the Treatment of Hepatocellular Carcinoma: Recent Advances in Molecular Mechanism and Drug Targeting. Curr Drug Targets 2025; 26:167-187. [PMID: 39385414 DOI: 10.2174/0113894501312571240920070441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/17/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024]
Abstract
The second largest cause of cancer-related death worldwide, Hepatocellular Carcinoma (HCC) is also the most common primary liver cancer. HCC typically arises in patients with liver cirrhosis. Existing synthetic medicines for treating chronic liver disease are ineffective and come with undesirable side effects. Although herbal remedies have widespread popularity, there is still a long road ahead before they are fully accepted by the scientific community. Secondary metabolites and phytochemicals found in plants are abundant in both the human diet and the non-human environment. Natural plant chemicals have been shown to be beneficial as therapeutic and chemopreventive treatments for a wide variety of chronic disorders. Many diseases, including HCC, can be effectively treated with the help of phytochemicals found in food. Resveratrol, curcumin, urolithin A, silibinin, quercetin, N-trans-feruloyl octopamine, emodin, lycopene, caffeine, and phloretin are all examples. Approximately, 60% of all anticancer medications are determined to be derived from natural substances, according to recent studies. Plant derivatives have played an important role in cancer due to their capacity to scavenge free radicals, limit cell proliferation, and set off apoptosis. The progression of HCC is linked to inflammatory signaling pathways, and this study sought to look at how novel approaches, such as phytomedicines, are being used to fight cancer. Recent advancements in molecular mechanisms and drug targeting for HCC have been discussed in this review.
Collapse
Affiliation(s)
| | - Ram Kumar Sahu
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Chauras Campus, Tehri Garhwal-249161, Uttarakhand, India
| | - Amit Roy
- Chhatrapati Shivaji Institute of Pharmacy, Bhilai, Chhattisgarh-491001, India
| |
Collapse
|
144
|
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
|
145
|
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
|
146
|
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
|
147
|
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
|
148
|
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
|
149
|
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
|
150
|
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
|