151
|
Hajim WI, Zainudin S, Daud KM, Alheeti K. Golden eagle optimized CONV-LSTM and non-negativity-constrained autoencoder to support spatial and temporal features in cancer drug response prediction. PeerJ Comput Sci 2024; 10:e2520. [PMID: 39896419 PMCID: PMC11784781 DOI: 10.7717/peerj-cs.2520] [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: 06/19/2024] [Accepted: 10/25/2024] [Indexed: 02/04/2025]
Abstract
Advanced machine learning (ML) and deep learning (DL) methods have recently been utilized in Drug Response Prediction (DRP), and these models use the details from genomic profiles, such as extensive drug screening data and cell line data, to predict the response of drugs. Comparatively, the DL-based prediction approaches provided better learning of such features. However, prior knowledge, like pathway data, is sometimes discarded as irrelevant since the drug response datasets are multidimensional and noisy. Optimized feature learning and extraction processes are suggested to handle this problem. First, the noise and class imbalance problems must be tackled to avoid low identification accuracy, long prediction times, and poor applicability. This article aims to apply the Non-Negativity-Constrained Auto Encoder (NNCAE) network to tackle these issues, enhance the adaptive search for the optimal size of sliding windows, and ensure that deep network architectures are adept at learning the vital hidden features. NNCAE methodology is used after performing the standard pre-processing procedures to handle the noise and class imbalance problem. This class balanced and noise-removed input data features are learned to train the proposed hybrid classifier. The classification model, Golden Eagle Optimization-based Convolutional Long Short-Term Memory neural networks (GEO-Conv-LSTM), is assembled by integrating Convolutional Neural Network CNN and LSTM models, with parameter tuning performed by the GEO algorithm. Evaluations are conducted on two large datasets from the Genomics of Drug Sensitivity in Cancer (GDSC) repository, and the proposed NNCAE-GEO-Conv-LSTM-based approach has achieved 96.99% and 97.79% accuracies, respectively, with reduced processing time and error rate for the DRP problem.
Collapse
Affiliation(s)
- Wesam Ibrahim Hajim
- Department of Applied Geology, College of Sciences, University of Tikrit, Tikrit, Salah ad Din, Iraq
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Suhaila Zainudin
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Kauthar Mohd Daud
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Khattab Alheeti
- Department of Computer Networking Systems College of Computer Sciences and Information Technology, University of Anbar, Ramadi, Al Anbar, Iraq
| |
Collapse
|
152
|
Peng Y, Yang X, Liu Y, Zhou J, Guo J, Ma B, Bai Y, Wu J, Hu D. The regulation of the cell cycle and epithelial-mesenchymal transition through FUCA2/GGH signaling promotes the progression of lung adenocarcinoma. Gene 2024; 939:149183. [PMID: 39710011 DOI: 10.1016/j.gene.2024.149183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/14/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The development of lung adenocarcinoma (LUAD) is intricately linked with cell cycle regulation and epithelial-mesenchymal transition (EMT). Our study, leveraging bioinformatics and database analysis, identified FUCA2 as a key gene influencing the prognosis and progression of LUAD. We observed that FUCA2 is highly expressed in LUAD and correlates with poor outcomes. Functionally, we assessed the role of this gene through cell cloning, scratch assays, transwell migration, and western blotting, revealing that FUCA2 knockdown significantly inhibits tumor cell proliferation and migration, downregulates the expression of cell cycle and EMT-related proteins, and markedly reduces tumor burden. Mechanistically, pathway enrichment analysis identified GGH as a downstream target of FUCA2. Knockdown of GGH similarly inhibits the proliferation, migration, and cell cycle progression of LUAD cells. FUCA2 upregulates GGH to modulate cell cycle and EMT in LUAD. Collectively, our findings indicate that the FUCA2/GGH axis promotes LUAD progression by regulating cell cycle and EMT.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Xingyu Yang
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Yafeng Liu
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jiawei Zhou
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Jianqiang Guo
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Bingfeng Ma
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China
| | - Ying Bai
- Department of Immunology, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Anhui Province Engineering Laboratory of Occupational Health and Safety, School of Medicine, Anhui University of Science and Technology, Huainan City, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan City, China.
| | - Dong Hu
- The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital), School of Medicine, Huainan City, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei City, China.
| |
Collapse
|
153
|
Louis D, Rizkalla CMZ, Rashad A. Cubosomes as Delivery System to Repositioning Nitrofurantoin in Breast Cancer Management. Drug Des Devel Ther 2024; 18:6173-6184. [PMID: 39722678 PMCID: PMC11668685 DOI: 10.2147/dddt.s499068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose Nitrofurantoin (NITRO), a long-standing antibiotic to treat urinary tract infections, is activated by Nitro reductases. This activation mechanism has led to its exploration for repositioning applications in controlling and treating breast cancer, which express a Nitro reductase gene. Methods NITRO Cubosomes were developed using hot homogenization according to 23-full factorial design. The factors studied included the ratio of drug to oily phase (1:10 and 2:10), the ratio of oily to aqueous phase (1:10 and 1:5), and the ratio of Glyceryl mono-oleate (GMO) to Poloxamer 407 (PX407) (0.25:1 and 0.5:1). A total of 8 systems were proposed and evaluated by measuring particle size, zeta potential, polydispersity index, and percentage of entrapment efficiency. Results S6 (1:10 drug: oily phase, 1:5 oily: aqueous phase and 0.5:1 GMO: PX407) with particle size 45.5 ±c1.1 nm and an entrapment efficiency of 98.6 ± 1.8% exhibited highest desirability and was selected for further analysis. The morphology of S6 was examined using TEM microscopy. The activation of NITRO from S6 reflected on intracellular viability of MCF-7 breast cancer cell line was investigated by an MTT assay. The findings indicated that S6 had the lowest IC50 value (83.99 ± 0.15 μg g/mL) compared to Free NITRO (174.54 ± 1.36 μg g/mL), suggesting enhanced efficacy compared to free NITRO. Conclusion Nitrofurantoin cubosomes can be candidates for repositioning in breast cancer management after encouraging further stability and in-vivo studies.
Collapse
Affiliation(s)
- Dina Louis
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | | | - Amira Rashad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
154
|
St-Laurent MP, Bochner B, Catto J, Davies BJ, Fankhauser CD, Garg T, Hamilton-Reeves J, Master V, Jensen BT, Lauridsen SV, Wulff-Burchfield E, Psutka SP. Increasing Life Expectancy in Patients with Genitourinary Malignancies: Impact of Treatment Burden on Disease Management and Quality of Life. Eur Urol 2024:S0302-2838(24)02746-5. [PMID: 39706786 DOI: 10.1016/j.eururo.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/24/2024] [Accepted: 11/24/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Treatment burden refers to the overall impact of medical treatments on a patient's well-being and daily life. Our objective is to evaluate the impact of treatment burden on quality of life (QoL) in patients with genitourinary (GU) malignancies, highlighting the importance of patient-reported outcomes (PROs) in clinical trials to inform treatment decisions and improve patient care. METHODS We conducted a narrative review of clinical trials focused on GU malignancy (prostate, bladder, and kidney) between January 2000 and June 2024, analyzing related PROs and findings regarding treatment burden. KEY FINDINGS AND LIMITATIONS Recent landmark clinical trials demonstrate significant improvements in overall survival across GU malignancies with novel therapies. However, the reporting of QoL outcomes in these trials is often inadequate, with many lacking comprehensive data or long-term impact. Current publications are increasingly evaluating treatment burden and its impact on patient well-being as a critical outcome, but most clinical trials to date have failed to assess treatment burden across key domains including financial, time and travel, and medication management. CONCLUSIONS AND CLINICAL IMPLICATIONS While advancements in treatment have extended longevity in patients with GU malignancies, the treatment burden associated with the receipt of novel agents and its implications for QoL remain inadequately uncharacterized.
Collapse
Affiliation(s)
- Marie-Pier St-Laurent
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Bernard Bochner
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James Catto
- Department of Urology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Benjamin J Davies
- Department of Urology Division of Health Services Research University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Tullika Garg
- Department of Urology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jill Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Viraj Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bente T Jensen
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Susanne V Lauridsen
- WHO-CC/Clinical Health Promotion Centre, the Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark; Center for Perioperative Optimization, Department of Surgery, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elizabeth Wulff-Burchfield
- Medical Oncology Division and Palliative Medicine Division, Department of Internal Medicine, University of Kansas School of Medicine, University of Kansas Cancer Center, The University of Kansas Health System, Kansas City, KS, USA
| | - Sarah P Psutka
- Department of Urology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
155
|
Ravichandiran P, Martyna A, Kochanowicz E, Maroli N, Kubiński K, Masłyk M, Boguszewska-Czubara A, Ramesh T. In Vitro and In Vivo Biological Evaluation of Novel 1,4-Naphthoquinone Derivatives as Potential Anticancer Agents. ChemMedChem 2024; 19:e202400495. [PMID: 39136593 DOI: 10.1002/cmdc.202400495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/13/2024] [Indexed: 10/16/2024]
Abstract
A novel library of naphthoquinone derivatives (3-5 aa) was synthesized and evaluated for their anticancer properties. Specifically, compounds 5 i, 5 l, 5 o, 5 q, 5 r, 5 s, 5 t, and 5 v demonstrated superior cytotoxic activity against the cancer cell lines that were studied. All the studied compounds exhibited a higher selectivity index (SI) and a favourable safety profile than the standard drug doxorubicin. Notably, compound 5 v displayed a greater cytotoxic effect on MCF-7 cells (IC50=1.2 μM, and 0.9 μM at 24 h and 48 h, respectively) compared to the standard drug doxorubicin (IC50=2.4 μM, and 2.1 μM at 24 h and 48 h, respectively). To further investigate the mechanism of cytotoxic effect, additional anticancer studies were conducted with 5 v in MCF-7 cells. The studies are including morphological changes, AO/EB (acridine orange/ethidium bromide) double staining, apoptosis analysis, cell colony assay, SDS-PAGE and Western blotting, cell cycle analysis, and detecting reactive oxygen species (ROS) assay. The findings showed that 5 v triggered cytotoxic effects in MCF-7 cells through the initiation of cell cycle arrest at the G1/S phase and necrosis. In vivo ecotoxicity studies indicated that 5 v had lower toxicity towards zebrafish larvae (LC50=50.15 μM) and had an insignificant impact on cardiac functions. In vivo xenotransplantation of MCF-7 cells in zebrafish larvae demonstrated a significant reduction in tumour volume in the xenograft. Approximately 95 % of the zebrafish larvae with 5 v xenografts survived after 10 days of the treatment. Finally, a computational modelling study was conducted on four protein receptors, namely ER, EFGR, BRCA1, and VEFGR2. The findings highlight the importance of the aminonaphthoquinone moiety, amide linkage, and propyl thio moiety in enhancing the anticancer properties. 5 v exhibited superior drug-likeness features and docking scores (-9.1, -7.1, -8.9, and -10.9 kcal/mol) compared to doxorubicin (-7.2, -6.1, -6.9, and -7.3 kcal/mol) against ER, EFGR, BRCA1, and VEGFR2 receptors, respectively. Therefore, the notable antitumor effects of naphthoquinone derivatives (3-5 aa) suggest that these molecular frameworks may play a role in the development of promising anticancer agents for cancer treatment.
Collapse
Affiliation(s)
- Palanisamy Ravichandiran
- R&D Education Center for Whole Life Cycle R&D of Fuel Cell Systems, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Department of Energy Storage/Conversion Engineering of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Present Address: Analytical, HP Green R & D Centre, Hindustan Petroleum Corporation Limited, KIADB Industrial Area, Devangundi, Hoskote, Bengaluru, Karnataka, 562114, India
| | - Aleksandra Martyna
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Elżbieta Kochanowicz
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Nikhil Maroli
- Department of Physics and Astronomy, University of Delaware, Newark, DE, 19716, USA
| | - Konrad Kubiński
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Ul. Chodźki 4 A, 20-093, Lublin, Poland
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
156
|
Maldonado-García JL, Fragozo A, Pavón L. Cytokine release syndrome induced by anti-programmed death-1 treatment in a psoriasis patient: A dark side of immune checkpoint inhibitors. World J Clin Cases 2024; 12:6782-6790. [PMID: 39687650 PMCID: PMC11525914 DOI: 10.12998/wjcc.v12.i35.6782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/24/2024] Open
Abstract
In recent years, cancer immunotherapy has introduced novel treatments, such as monoclonal antibodies, which have facilitated targeted therapies against tumor cells. Programmed death-1 (PD-1) is an immune checkpoint expressed in T cells that regulates the immune system's activity to prevent over-activation and tissue damage caused by inflammation. However, PD-1 is also expressed in tumor cells and functions as an immune evasion mechanism, making it a therapeutic target to enhance the immune response and eliminate tumor cells. Consequently, immune checkpoint inhibitors (ICIs) have emerged as an option for certain tumor types. Nevertheless, blocking immune checkpoints can lead to immune-related adverse events (irAEs), such as psoriasis and cytokine release syndrome (CRS), as exemplified in the clinical case presented by Zhou et al involving a patient with advanced gastric cancer who received sintilimab, a monoclonal antibody targeting PD-1. Subsequently, the patient experienced exacerbation of psoriasis and CRS. The objective of this editorial article is to elucidate potential immunologic mechanisms that may contribute to the development of CRS and psoriasis in patients receiving ICIs. It is crucial to acknowledge that while ICIs offer superior safety and efficacy compared to conventional therapies, they can also manifest irAEs affecting the skin, gastrointestinal tract, or respiratory system. In severe cases, these irAEs can lead to life-threatening complications such as circulatory shock or multiorgan failure. Consequently, it is recommended that patients receiving ICIs undergo regular monitoring to identify and manage these adverse events effectively.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán 04510, Ciudad de México, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Mexico City 1134, Ciudad de México, Mexico
| | - Ana Fragozo
- Unidad de Desarrollo e Investigación en Bioterapéuticos, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Ciudad de México, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 11340, Mexico
| |
Collapse
|
157
|
Madikonda AK, Ajayakumar A, Nadendla S, Banothu J, Muripiti V. Esterase-responsive nanoparticles (ERN): A targeted approach for drug/gene delivery exploits. Bioorg Med Chem 2024; 116:118001. [PMID: 39556942 DOI: 10.1016/j.bmc.2024.118001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Nanoparticles are being developed to enhance drug delivery to cancer tumors, leveraging advantages such as the enhanced permeability and retention (EPR) effect. However, traditional nanoparticles often face challenges with low specificity for cancer cells, leading to inefficient delivery and unwanted side effects. Esterase-responsive nanoparticles offer a maximum targeted approach to tumor cells because they release their therapeutic payload at the tumor site under the influence of esterase activity. This review explores the role of esterase-responsive nanoparticles in drug and gene delivery, examines esterase prodrug therapy, and discusses prostate-specific membrane antigen (PSMA) targets esterase-responsive nanoparticles in prostate cancer treatment. Additionally, we reviewed the current research progress and future potential of esterase-responsive nanoparticles in enhancing drug and gene delivery.
Collapse
Affiliation(s)
- Ashok Kumar Madikonda
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Amritha Ajayakumar
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Sudeena Nadendla
- Department of Chemistry, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Janardhan Banothu
- Department of Chemistry, National Institute of Technology Calicut, Kozhikode 673601, Kerala, India
| | - Venkanna Muripiti
- Department of Education, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India.
| |
Collapse
|
158
|
Khaled NA, Ibrahim MA, Mohamed NA, Ahmed SA, Ahmed NS. DFT studies on N-(1-(2-bromobenzoyl)-4-cyano-1H-pyrazol-5-yl). SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 323:124864. [PMID: 39067358 DOI: 10.1016/j.saa.2024.124864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/01/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
In this work, molecular descriptors of N-(1-(2-bromobenzoyl)-4-cyano-1H-pyrazol-5-yl) halogenated benzamides (1a-h) have been computed using a quantum chemical technique through DFT. Prior work involved the synthesis of compounds (1a-h) and the assessment of their anticancer activity on breast, colon, and liver tumors: MCF-7, HCT-116, and HepG-2 cell lines respectively. Since 1a, 1b, and 1d showed the most potential anticancer impact, their ability to inhibit EGFRWT was investigated. Based on the biological data, 1b inhibited EGFRWT the most. According to the docking evaluation, an H-bond with the threonine residue was one of the main non-covalent contacts between 1b and the EGFRWT active site residues. PES, MESP, HOMOs, LUMOs, energy band gap, global reactivity indices [electron affinity (A), ionization energies (I), electrophilicity index (ω), nucleophilicity index (ε), chemical potential (μ), electronegativity (χ), hardness (η), and softness (S)], condensed Fukui functions, NBO, and NCIs are the molecular descriptors of 1a-h that were computed using DFT technique. According to the theoretical investigation results, compounds (1a-h) might have anticancer effects; these findings are consistent with the biological findings from our previous research. Compound 1b had the lowest binding energy, according to an assessment of the binding energies between the threonine and the three most active compounds (1a, 1b, and 1d). This is consistent with the outcomes of the docking study and the biological examination of the influence of 1a, 1b, and 1d on EGFRWT.
Collapse
Affiliation(s)
- Nada A Khaled
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Medhat A Ibrahim
- Spectroscopy Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt; Molecular Modeling and Spectroscopy Laboratory, Centre of Excellence for Advanced Science, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt.
| | - Neama A Mohamed
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Sayed A Ahmed
- Department of Chemistry, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt; Basic Science Department, Faculty of Engineering, Nahda University Beni-Suef (NUB), Beni Suef, Egypt
| | - Nesreen S Ahmed
- Therapeutical Chemistry Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
159
|
Bidram M, Ganjalikhany MR. Bioactive peptides from food science to pharmaceutical industries: Their mechanism of action, potential role in cancer treatment and available resources. Heliyon 2024; 10:e40563. [PMID: 39654719 PMCID: PMC11626046 DOI: 10.1016/j.heliyon.2024.e40563] [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: 02/08/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer is known as the main cause of mortality in the world, and every year, the rate of incidence and death due to cancer is increasing. Bioactive peptides are one of the novel therapeutic options that are considered a suitable alternative to toxic chemotherapy drugs because they limit side effects with their specific function. In fact, bioactive peptides are short amino acid sequences that obtain diverse physiological functions to maintain human health after being released from parent proteins. This group of biological molecules that can be isolated from different types of natural protein sources has attracted much attention in the field of pharmaceutical and functional foods production. The current article describes the therapeutic benefits of bioactive peptides and specifically and extensively reviews their role in cancer treatment, available sources for discovering anticancer peptides, mechanisms of action, production methods, and existing challenges.
Collapse
Affiliation(s)
- Maryam Bidram
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
160
|
Valmonte DJ, Quartey NK, Gutierrez F, Mendoza J, Papadakos J, Giuliani M. Education and Training Needs of Health Care Professionals in the Philippines Encountering Patients with Lung Oligometastatic Cancers. Curr Oncol 2024; 31:7950-7959. [PMID: 39727709 DOI: 10.3390/curroncol31120586] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
This study aimed to examine the education and training needs of health care practitioners (HCPs) in the Philippines who encounter lung oligometastatic cancer patients. Lung oligometastatic disease is among the most common sites for cancer spread and has the most established practices for treating oligometastases. A modified version of the Hennessy-Hicks Training Needs Assessment Questionnaire was administered online to HCPs working in private and public centers in the Philippines. HCPs were recruited via purposive sampling. Twenty-seven HCPs completed the questionnaire (47% response rate). Respondents were mostly female (59%) and between the ages of 30 and 39 years (70%). Three-quarters (74%) were consultants, and most respondents were radiation oncologists (44%) or medical oncologists (30%). Medical oncologists rated Management/Supervisory Tasks (mean = 1.42) as their highest area of training need while radiation oncologists rated Clinical Tasks (mean = 1.30) as their highest training need. Pulmonologists (mean = 0.60) and other specialists (mean = 1.00) rated Administration tasks as their top area of training need. The clinical task-related category was rated the highest need among the continuing medical education topics. This study provides valuable insights for the implementation and advancement of a comprehensive curriculum in clinical oncology, specifically designed to enhance the administrative, clinical, and research capacities of oncologists who encounter oligometastatic lung disease in the Philippines.
Collapse
Affiliation(s)
- Daphne J Valmonte
- Department of Radiation Oncology, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
| | - Naa Kwarley Quartey
- Cancer Education, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
| | - Fatima Gutierrez
- MakatiMed Wellness Center, 7th Floor, Ayala North Exchange Tower 1 6796 Ayala Avenue, cor Salcedo, Legaspi Village, Makati 1226, Philippines
| | - Janel Mendoza
- MakatiMed Wellness Center, 7th Floor, Ayala North Exchange Tower 1 6796 Ayala Avenue, cor Salcedo, Legaspi Village, Makati 1226, Philippines
| | - Janet Papadakos
- Cancer Education, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- The Institute for Education Research, University Health Network, 222 St. Patrick Street, Toronto, ON M5T 1V4, Canada
- The Institute for Health Policy, Management & Evaluation, University of Toronto, 155 College Street, Suite 425, Toronto, ON M5T 3M6, Canada
| | - Meredith Giuliani
- Department of Radiation Oncology, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- Cancer Education, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- The Institute for Education Research, University Health Network, 222 St. Patrick Street, Toronto, ON M5T 1V4, Canada
- Department of Radiation Oncology, University of Toronto, 149 College Street, Suite 504, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
161
|
Munteanu A, Gogulescu A, Șoica C, Mioc A, Mioc M, Milan A, Lukinich-Gruia AT, Pricop MA, Jianu C, Banciu C, Racoviceanu R. In Vitro and In Silico Evaluation of Syzygium aromaticum Essential Oil: Effects on Mitochondrial Function and Cytotoxic Potential Against Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2024; 13:3443. [PMID: 39683236 DOI: 10.3390/plants13233443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The current study proposes the in vitro and in silico anticancer evaluation of clove (Syzygium aromaticum L.) essential oil (CEO). The steam hydrodistillation method used yielded 10.7% (wt) CEO. GC-MS analysis revealed that the obtained oil is rich in eugenol (75%), β-caryophyllene (20%), and α- caryophyllene (2.8%) and also contains several other minor components accounting for approximately 1.5%. The DPPH-based scavenging antioxidant activity was assessed for the obtained CEO, exhibiting an IC50 value of 158 μg/mL. The cytotoxic effects of CEO, its major component eugenol, and CEO solubilized with Tween-20 and PEG-400 were tested against both noncancerous HaCaT cells and HT-29 human colorectal adenocarcinoma, RPMI-7951 melanoma, A431 skin carcinoma, and NCI-H460 non-small lung cancer cells, using the Alamar Blue and LDH assay after 48 h treatment. The Tween-20 and PEG-400 CEO formulations, at 200 μg/mL, recorded the highest cytotoxic and selective effects against RPMI-7951 (72.75% and 71.56%), HT-29 (71.51% and 45.43%), and A431 cells (61.62% and 59.65%). Furthermore, CEO disrupted mitochondrial function and uncoupled oxidative phosphorylation. This effect was more potent for the CEO against the RPMI-7951 and HT-29 cells, whereas for the other two tested cell lines, a more potent inhibition of mitochondrial function was attributed to eugenol. The present study is the first to specifically investigate the effects of CEO and Tween-20 and PEG-400 CEO formulations on the mitochondrial function of RPMI-7951, HT-29, A431, and NCI-H460 cancer cell lines using high-resolution respirometry, providing novel insights into their impact on mitochondrial respiration and bioenergetics in cancer cells. The results obtained may explain the increased ROS production observed in cancer cell lines treated with eugenol and CEO. Molecular docking identified potential protein targets, related to the CEO anticancer activity, in the form of PI3Kα, where the highest active theoretical inhibitor was calamenene (-7.5 kcal/mol). Docking results also showed that calamenene was the overall most active theoretical inhibitor for all docked proteins and indicated a potential presence of synergistic effects among all CEO constituents.
Collapse
Affiliation(s)
- Andreea Munteanu
- Department of Internal Medicine IV, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Armand Gogulescu
- Department XVI: Balneology, Medical Rehabilitation and Rheumatology, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Codruța Șoica
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Alexandra Mioc
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Marius Mioc
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Andreea Milan
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | | | - Maria-Alexandra Pricop
- OncoGen Centre, Clinical County Hospital "Pius Branzeu", Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania
- Department of Applied Chemistry and Environmental Engineering and Inorganic Compounds, Faculty of Industrial Chemistry, Biotechnology and Environmental Engineering, Polytechnic University of Timisoara, Vasile Pârvan 6, 300223 Timisoara, Romania
| | - Calin Jianu
- Faculty of Food Engineering, Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania"Timisoara, Calea Aradului 119, 300645 Timisoara, Romania
| | - Christian Banciu
- Department of Internal Medicine IV, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania
| | - Roxana Racoviceanu
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| |
Collapse
|
162
|
Cecchetti S, Federici C, Canese R, Iorio E, Huber V, Pisanu ME, Chirico M, Iessi E, Camerini S, Casella M, Matteucci A, Macchia D, Spada M, Lugini L. NK cells-derived extracellular vesicles potency in the B cell lymphoma biotherapy. Front Immunol 2024; 15:1503857. [PMID: 39712029 PMCID: PMC11659271 DOI: 10.3389/fimmu.2024.1503857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Extracellular vesicles of Natural Killer cells (NKEV) exert an antitumor effect towards hematopoietic and solid tumors and have an immune modulating effect, suggesting a promising role in immune and biotherapy. In this study, a continuation of our former works, we demonstrated a network by mass spectrometry analysis between NKEV protein cargo and antitumor effects. Human healthy NKEV, both exosomes and microvesicles, have a significant and direct cytotoxic effect against human B cell lymphoma in in vitro and in vivo conditions. Methods We isolated extracellular vesicles from in vitro amplified healthy human NK cells and their treatment efficacy was monitored by cytometry analyses, in vivo MRI/MRS measurements, ex vivo MRS analyses and immunohistochemistry. Results We observed a remarkable NKEV cytotoxic effect, mainly by apoptosis, on B cell lymphoma in vitro when exosomes and microvesicles were administered simultaneously. In vivo results showed metabolic alterations in SCID mice xenografts after NKEV treatment, associated with a significant reduction of tumor growth (64%). In the in vivo 1H MR spectra we found a significant increase in the tumor lipid/lactate and in taurine signals, both considered as apotosis markers. Ex vivo lymphoma metabolomics revealed a significant increase in fatty acid (FA) pool and decrease in unsaturated and mono-unsaturated FA in treated groups, as compared to control one, thus suggesting an alteration of tumor homeostasis. Immunohistochemistry analyses confirmed the reduction of B-cell lymphoma proliferation rate, as well as the induction of apoptosis following the NKEV treatment. Conclusions This study underscore the importance of NKEV as a novel biological acellular tool for B-cell lymphoma treatment, probably having a greater effect on combined treatment regimens. These nanovesicles have an extraordinary potential in innovative cancer immunotherapy, representing a safe and efficient tool naturally circulating in healthy individuals and ready to maintain the immune homeostasis, and therefore a good organism healthy state.
Collapse
Affiliation(s)
- Serena Cecchetti
- Core Facilities, Confocal Microscopy Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Federici
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rossella Canese
- Core Facilities, MRI and HR-NMR Units, Istituto Superiore di Sanità, Rome, Italy
| | - Egidio Iorio
- Core Facilities, MRI and HR-NMR Units, Istituto Superiore di Sanità, Rome, Italy
| | - Veronica Huber
- Unit of Immunotherapy of human tumors, Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Elena Pisanu
- Core Facilities, MRI and HR-NMR Units, Istituto Superiore di Sanità, Rome, Italy
| | - Mattea Chirico
- Core Facilities, MRI and HR-NMR Units, Istituto Superiore di Sanità, Rome, Italy
| | - Elisabetta Iessi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Serena Camerini
- Core Facilities, Mass Spectrometry Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Marialuisa Casella
- Core Facilities, Mass Spectrometry Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Matteucci
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- Centre for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Centre for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Luana Lugini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
163
|
Khalili-Hezarjaribi H, Bahrami AR, Sh Saljooghi A, Matin MM. Modified mesoporous silica nanocarriers containing superparamagnetic iron oxide nanoparticle, 5-fluorouracil or oxaliplatin, and metformin as a radiosensitizer, significantly impact colorectal cancer radiation therapy. Int J Pharm 2024; 666:124838. [PMID: 39419365 DOI: 10.1016/j.ijpharm.2024.124838] [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: 06/21/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024]
Abstract
This study investigates the anticancer effects of SPION-based silica nanoparticles carrying 5-fluorouracil (5-FU) or oxaliplatin (OX), and metformin (MET) on colorectal cancer cells. Nanocarriers were equipped with pH-responsive gold gatekeepers for controlled release, PEGylation for longer circulation, and folic acid (FA) for targeted delivery. The effects were evaluated by investigating cell viability, cellular uptake, flow cytometry, and clonogenic assay in vitro. The efficacy of the system was also tested in vivo on C57BL/6 mice bearing HT-29 tumors, and potential side effects were evaluated. Nanocarriers were synthesized with hydrodynamic diameters of 79.8 nm for 5-FU and 85.2 nm for OX; zeta potentials of -21 and -22 mV, respectively, and remained stable after 72 h. Encapsulation efficiencies were 85 % for 5-FU, 80 % for OX, and 83 % for MET, with loading capacities of 44 %, 38 %, and 41 %, respectively. Drug release in acidic buffer was 38.7 % for 5-FU, 32.8 % for OX, and 43.5 % for MET. MTT assay showed increased toxicity due to FA conjugation, while PEGylation reduced the hemolysis activity. Targeted nanocarriers demonstrated superior cellular uptake and tumor localization compared to non-targeted variants. The combination of 5-FU-MET and OX-MET nanocarriers with radiation therapy (RT) demonstrated the greatest effect on their antitumor activity, accompanied by minimal side effects indicating effective tumor targeting in vivo. MRI and CT imaging further supported these findings. This study underscores the synergistic impact of MET alongside RT on the inhibition of cancer cells and tumor growth for both targeted 5-FU and OX nanocarriers reflecting the significant radiosensitizing properties of MET.
Collapse
Affiliation(s)
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
164
|
Zamani M, Sadeghi E, Mokarram P, Kadkhodaei B, Ghasemi H. Autophagy related proteins as potential biomarkers in predicting cancer prognosis after chemoradiotherapy: systematic review and meta-analysis. Int J Radiat Biol 2024; 101:232-239. [PMID: 39625853 DOI: 10.1080/09553002.2024.2435320] [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: 06/29/2024] [Revised: 10/30/2024] [Accepted: 11/24/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND Resistance to chemo- and radiotherapy is the main obstacle in cancer treatment success, which results in cancer's poor prognosis. Therefore finding the exact mechanism of resistance may contribute to addressing this concern. This could result in improved cancer prognosis and survival outcomes for cancer patients by targeting the basic causes of resistance. AIM This systematic review and meta-analysis assessed the potential of using autophagy-related proteins as prognostic biomarkers in radiotherapy-treated patients. METHODS Following PRISMA guidelines, we systematically reviewed 956 studies from PubMed, Scopus, and Web of Science databases until April 2023. The keywords used for this purpose were 'cancer', 'radiotherapy', 'prognosis', and 'Autophagy'. Then the related meta-analysis was performed using STATA software. RESULTS Four studies met the inclusion criteria. Upregulation of autophagy markers (LC3B, Beclin1 and ULK1) and subsequent activation of autophagy were significantly associated with a higher risk of mortality (1.95 times) in radiotherapy-treated groups compared with patients with low expression of these markers. Although such results were observed for recurrence-free survival (RFS); however, it was not significant. CONCLUSION The findings of this meta-analysis suggest that autophagy activation may be a critical factor in resistance to radiotherapy and subsequent poor survival rates in cancer patients. Consequently, assessing the expression of autophagy-related markers like Beclin1, LC3II, P62, and ULK may be a useful method for monitoring cancer prognosis following radiotherapy.
Collapse
Affiliation(s)
- Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Erfan Sadeghi
- Department of Biostatistics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Department of Biochemistry Shiraz, University of Medical Sciences, Shiraz, Iran
| | - Behnam Kadkhodaei
- Department of Radiation Oncology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Ghasemi
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
165
|
Kim CG, Jose J, Hay MP, Choi PJ. Novel Prodrug Strategies for the Treatment of Tuberculosis. Chem Asian J 2024; 19:e202400944. [PMID: 39179514 PMCID: PMC11613820 DOI: 10.1002/asia.202400944] [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: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
The emergence of drug-resistant strains of Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis, is on the rise and increasing antimicrobial resistance is a global threat. This phenomenon necessitates new drug design methods such as a prodrug strategy to develop novel antitubercular agents. The prodrug strategy is a viable and useful means to improve the absorption, distribution, metabolism, excretion and toxicity (ADMET) profiles of pharmacologically active agents. Granulomas are a pathological hallmark of M.tb infection and bear a remarkable resemblance to the tumour microenvironment, including regions of hypoxia. The hypoxic environment observed in the two structures offer an exceptional opportunity to deliver antitubercular agents selectively in a similar manner to hypoxia activated prodrugs in cancer therapy. Nitroimidazoles have been studied extensively as bioactivated prodrugs of cancer, and their suitability as substrates for mammalian reductases highlight their huge potential. This review will discuss the mechanism of action and resistance mechanisms of the current prodrugs used for the treatment of tuberculosis. It will also highlight the potential advantages and challenges of using hypoxia activated prodrugs as a viable strategy to target latent M.tb in hypoxic regions of granulomas.
Collapse
Affiliation(s)
- Christine G. Kim
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| | - Peter J. Choi
- Auckland Cancer Society Research Centre, School of Medical SciencesUniversity of AucklandPrivate Bag 92019Auckland1142New Zealand
| |
Collapse
|
166
|
Kulshrestha S, Goel A, Banerjee S, Sharma R, Khan MR, Chen KT. Metabolomics and network pharmacology-guided analysis of TNF-α expression by Argemone mexicana (Linn) targeting NF-kB the signalling pathway in cancer cell lines. Front Oncol 2024; 14:1502819. [PMID: 39687882 PMCID: PMC11648424 DOI: 10.3389/fonc.2024.1502819] [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: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Cancer has emerged as one of the leading causes of fatality all over the world. Phytoconstituents are being studied for their synergistic effects, which include disease prevention by altering molecular pathways and immunomodulation without side effects. The present experiment aims to explore the cancer preventive activities of Argemone mexicana Linn leaves extract in skin cancer cell lines (A431) and colon cancer cell lines (COLO 320DM)). In addition, TNF-α expression patterns and NF-kB signaling pathways have been examined. Methods LC/MS study of Argemone mexicana Linn extracts in various solvents revealed anti-cancerous phytoconstituents. Network pharmacology analysis used Binding DB, STRING, DAVID, and KEGG for data mining to evaluate predicted compounds using functional annotation analysis. Cytoscape 3.2.1 created "neighbourhood approach" and networks. The MNTD of these extracts was tested on L929 fibroblasts. Skin cancer (A431) and colon cancer (COLO 320DM) cell lines were tested for IC50 inhibition. Evaluation of TNF-α and NF-kB expression in cell culture supernatants and homogenates reveals anti-cancerous effects. Results LC-MS analysis of extracts predicted the presence of anticancer alkaloids Berberine, Atropine, Argemexicin, and Argemonin. In Network pharmacology analysis, enrichment was linked to the PI3-AKT pathway for both cancer types. MNTD was calculated at 1000μg/ml in L929. The ethanolic extract at 1000μg/ml significantly inhibited skin cancer cell proliferation by 67% and colon cancer cells by 75%. Ethanolic extract significantly reduced TNF-α expression in both cell lines (p<0.001), with the highest inhibition at 1000μg/ml. In TNF-α stimulated cell lines, 1000μg/ml ethanolic extract significantly reduced the regulation of the NF-kB pathway, which plays a role in cancer progression (p<0.001). Conclusion Argemone mexicana Linn. known as 'swarnkshiri' in Ayurveda has been reported to be used by the traditional healers for the treatment of psoriasis and its anti-inflammatory and anti-cancerous properties, according to the Indian Medicinal Plant dictionary. In the experiment, the abatement in the expression of inflammatory cytokine TNF-α and inhibition of NF-kB transcription factor activation could be linked with the downregulation of cancer cell proliferation. The study revealed the anticancer activity of Argemone mexicana Linn in the cancer cell lines and paved a pathway for molecular approaches that could be explored more in In vivo studies.
Collapse
Affiliation(s)
| | - Anjana Goel
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Subhadip Banerjee
- School of Natural Products Studies, Jadavpur University, Kolkata, West Bengal, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| | - Mohammad Rashid Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Kow-Tong Chen
- Department of Occupational Medicine, Tainan Municipal Hospital (managed by ShowChwan Medical Care Corporation), Tainan, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
167
|
Lin Z, Assaraf YG, Kwok HF. Peptides for microbe-induced cancers: latest therapeutic strategies and their advanced technologies. Cancer Metastasis Rev 2024; 43:1315-1336. [PMID: 39008152 DOI: 10.1007/s10555-024-10197-4] [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: 02/26/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
Cancer is a significant global health concern associated with multiple distinct factors, including microbial and viral infections. Numerous studies have elucidated the role of microorganisms, such as Helicobacter pylori (H. pylori), as well as viruses for example human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), in the development of human malignancies. Substantial attention has been focused on the treatment of these microorganism- and virus-associated cancers, with promising outcomes observed in studies employing peptide-based therapies. The current paper provides an overview of microbe- and virus-induced cancers and their underlying molecular mechanisms. We discuss an assortment of peptide-based therapies which are currently being developed, including tumor-targeting peptides and microbial/viral peptide-based vaccines. We describe the major technological advancements that have been made in the design, screening, and delivery of peptides as anticancer agents. The primary focus of the current review is to provide insight into the latest research and development in this field and to provide a realistic glimpse into the future of peptide-based therapies for microbe- and virus-induced neoplasms.
Collapse
Affiliation(s)
- Ziqi Lin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Instituteof Technology, Haifa, 3200003, Israel
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
168
|
Darvishi S, Hosseinzadeh H, Kazeminava F, Mahoutforoush A, Tajik M, Rasoulzadehzali M, Mohammadi R, Sadjadi S, Javanbakht S. Heparin-functionalized Cu-based metal-organic framework: An efficient active and passive targeting nanocarrier for anticancer doxorubicin drug delivery. Int J Biol Macromol 2024; 282:136648. [PMID: 39437945 DOI: 10.1016/j.ijbiomac.2024.136648] [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: 07/06/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
In this study, by innovative combining the unique characteristics of Cu-based metal-organic framework (MOF) with the versatile attributes of saccharides (i.e., heparin, Hep), a promising approach is established for active and passive targeting DDS, Cu-MOF/Hep, with a pH-controlled release profile and enhanced drug efficacy. The characterization of the synthesized materials (i.e., FT-IR, XRD, SEM, EDX, TEM, DLS, and TGA) confirms the successful synthesis of Cu-MOF/Hep. In vitro studies concerning the loading and release of DOX observed that a higher amount of DOX was released at pH 5 (>90 % on 96 h, 41 °C) compared to pH 7.4 (<10 % on 96 h, 37 °C). The sensitive feature of the used MOF to the pH conditions increased the drug release in environmental conditions similar to cancer tissues. Furthermore, cytotoxicity assessments indicated notable cytotoxicity effects of DOX-loaded Cu-MOF/Hep on MCF-7 cells (IC50: ∼10 μg/mL in 48 h) with a significant apoptosis rate. The existence of CD44 receptors on the surfaces of cells underscores the significance of Hep-modified systems in facilitating the apoptosis of cancerous cells. The results suggest that the combined Cu-MOF and Hep have the potential to be a viable option for creating platforms that deliver anticancer treatments.
Collapse
Affiliation(s)
- Sima Darvishi
- Department of Chemistry, School of Physic and Chemistry, Alzahra University, PO Box 1993891176, Vanak, Tehran, Iran
| | - Hossein Hosseinzadeh
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Fahimeh Kazeminava
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Mahoutforoush
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Tajik
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, USA
| | | | - Reza Mohammadi
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Samaheh Sadjadi
- Gas Conversion Department, Faculty of Petrochemicals, Iran Polymer and Petrochemical Institute, PO Box 14975-112, Tehran, Iran.
| | - Siamak Javanbakht
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
169
|
Alotaibi B, A El-Masry T, Elekhnawy E, Mokhtar FA, El-Seadawy HM, A Negm W. Studying the effects of secondary metabolites isolated from Cycas thouarsii R.Br. leaves on MDA-MB-231 breast cancer cells. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:103-113. [PMID: 38279824 DOI: 10.1080/21691401.2024.2306529] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/03/2024] [Indexed: 01/29/2024]
Abstract
The various therapeutic drugs that are currently utilized for the management of cancer, especially breast cancer, are greatly challenged by the augmented resistance that is either acquired or de novo by the cancer cells owing to the long treatment periods. So, this study aimed at elucidating the possible anticancer potential of four compounds 7, 4', 7'', 4'''-tetra-O-methyl amentoflavone, hesperidin, ferulic acid, and chlorogenic acid that are isolated from Cycas thouarsii leaves n-butanol fraction for the first time. The MTT assay evaluated the cytotoxic action of four isolated compounds against MDA-MB-231 breast cancer cells and oral epithelial cells. Interestingly, ferulic acid revealed the lowest IC50 of 12.52 µg/mL against MDA-MB-231 cells and a high IC50 of 80.2 µg/mL against oral epithelial cells. Also, using an inverted microscope, the influence of ferulic acid was studied on the MDA-MB-231, which revealed the appearance of apoptosis characteristics like shrinkage of the cells and blebbing of the cell membrane. In addition, the flow cytometric analysis showed that the MDA-MB-231 cells stained with Annexin V/PI had a rise in the count of the cells in the early and late apoptosis stages. Moreover, gel electrophoresis detected DNA fragmentation in the ferulic acid-treated cells. Finally, the effect of the compound was tested at the molecular level by qRT-PCR. An upregulation of the pro-apoptotic genes (BAX and P53) and a downregulation of the anti-apoptotic gene (BCL-2) were observed. Consequently, our study demonstrated that these isolated compounds, especially ferulic acid, may be vital anticancer agents, particularly for breast cancer, through its induction of apoptosis through the P53-dependent pathway.
Collapse
Affiliation(s)
- Badriyah Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Tanta University, Tanta, Egypt
| | - Fatma A Mokhtar
- Department of Pharmacognosy, El Saleheya El Gadida University, Sharkia, Egypt
| | | | - Walaa A Negm
- Department of Pharmacognosy, Tanta University, Tanta, Egypt
| |
Collapse
|
170
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
171
|
Ashokkumar M, Palanisamy K, Ganesh Kumar A, Muthusamy C, Senthil Kumar KJ. Green synthesis of silver and copper nanoparticles and their composites using Ocimum sanctum leaf extract displayed enhanced antibacterial, antioxidant and anticancer potentials. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:438-448. [PMID: 39239690 DOI: 10.1080/21691401.2024.2399938] [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: 06/11/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Green-synthesized silver and copper nanoparticles (NPs), along with their composites, exhibit various biological activities. Ocimum sanctum (Holy basil), traditionally used as medicine in South Asia, treats respiratory disorders, digestive issues, skin diseases and inflammatory conditions. Modern scientific studies support these bioactivities; however, no studies have investigated their bioactivity in combination with NPs. In this study, silver and copper NPs were synthesized using AgNO3 and CuSO4·5H2O solutions, respectively, with Ocimum sanctum leaf extract, and their antibacterial, antioxidant and anticancer properties were examined. Spectroscopic analyses, including Fourier transform infra-red (FTIR), transmission electron microscopy (TEM) and X-ray diffraction (XRD), elucidated the physicochemical characteristics of the green-synthesized nanoparticles (Os-AgNPs and Os-CuNPs), revealing sizes of 11.7 and 13.1 nm, respectively. The Os-AgNPs:Os-CuNPs nano-composite with a 1:2 ratio exhibited a zone of inhibition ranging from 8 to 12 mm against tested bacterial pathogens. Additionally, the NPs and their composites demonstrated potent antioxidant activity, with notable 2-diphenyl-2-picrylhydrazyl (DPPH) scavenging activity observed in composites with ratios of 2:1 and 1:2. Furthermore, they displayed potential anticancer activity against human leukaemia (Jurkat) cancer cells. Although no distinct difference in anticancer property was observed among the NPs and their composites, our study highlights their well-defined nanostructure and significant biological activity, suggesting their potential as therapeutic agents in the pharmaceutical industry.
Collapse
Affiliation(s)
- M Ashokkumar
- Department of Physics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, India
| | - K Palanisamy
- Department of Chemistry, Srinivasan College of Arts and Science(Affiliated to Bharathidasan University, Tiruchirappalli), Perambalur, India
| | - A Ganesh Kumar
- Department of Microbiology, Centre for Research and Development, Hindustan College of Arts & Science, Kelambakkam, India
| | - C Muthusamy
- Department of Biotechnology, Srinivasan College of Arts and Science (Affiliated to Bharathidasan University, Tiruchirappalli), Perambalur, India
| | - K J Senthil Kumar
- Center for General Education, National Chung Hsing University, Taichung, Taiwan
- Bachelor Program of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
172
|
Hamid O, Hamidi N. Enhancing immuno-oncology efficacy with H1-antihistamine in cancer therapy: a review of current research and findings. Curr Med Res Opin 2024; 40:2139-2146. [PMID: 39503414 DOI: 10.1080/03007995.2024.2427323] [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: 08/01/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/11/2024]
Abstract
Cancer remains a major global cause of death, posing significant treatment challenges. The interactions between tumor cells and the tumor microenvironment (TME) are crucial in influencing tumor initiation, progression, metastasis, and treatment response. There has been significant research and clinical interest in targeting the TME as a therapeutic approach in cancer, with advancements being made through drug development. Histamine binds to HRH1 receptors on the TME, which inhibit CD8+ T cell activity, promote tumor growth, and contribute to resistance against immunotherapy. By inhibiting CD8+ T cells, the effectiveness of immunotherapies targeting these cells is reduced. By blocking the HRH1 pathway, H1-antihistamines can mitigate this suppression and enhance the response to immunotherapies that target CD8+ T cells. Therefore, understanding the role of histamine and its potential impact on T cells and the role of H1-antihistamines in improving immune-oncology (I/O) agents' efficacy ultimately could lead to more effective cancer therapies. The objective of this review is to examine the current literature to investigate the potential role of H1-antihistamines on the effectiveness of I/O drugs and their role in enhancing treatment against cancer. We conducted a comprehensive literature search, which included multiple databases including PubMed, Google Scholar, and EMBASE, as well as a search of oncology congresses. Our literature review initially identified thirty studies. Twenty-three of these were excluded for failing to meet inclusion criteria, which varied from study design to the type of antihistamines and patient populations involved. The clinical studies investigated the effect of different generations of H1-antihistamines in combination with I/O treatments on patients' outcomes. The findings from these studies indicated that patients using H1-antihistamines concomitantly with I/O agents experienced longer median overall survival (mOS), progression-free survival (mPFS), or improved survival compared to those who did not use antihistamines. Additionally, these trials differentiated between cationic and non-cationic H1-antihistamines, revealing that users of cationic antihistamines had overall better outcomes in terms of longer mOS and mPFS. The assessed trials were consistent in their comparisons of quantitative and qualitative, efficacy, and safety outcomes.
Collapse
Affiliation(s)
- Oday Hamid
- Department of Oncology, AstraZeneca/University of Michigan College of Pharmacy, Gaithersburg, MD, USA
| | - Negar Hamidi
- Department of Oncology, AstraZeneca/University of Maryland School of Pharmacy, Baltimore, MD, USA
| |
Collapse
|
173
|
Budhbaware T, Rathored J, Shende S. Molecular methods in cancer diagnostics: a short review. Ann Med 2024; 56:2353893. [PMID: 38753424 PMCID: PMC11100444 DOI: 10.1080/07853890.2024.2353893] [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: 02/14/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND One of the ailments with the greatest fatality rates in the 21st century is cancer. Globally, molecular methods are widely employed to treat cancer-related disorders, and the body of research on this subject is growing yearly. A thorough and critical summary of the data supporting molecular methods for illnesses linked to cancer is required. OBJECTIVE In order to guide clinical practice and future research, it is important to examine and summarize the systematic reviews (SRs) that evaluate the efficacy and safety of molecular methods for disorders associated to cancer. METHODS We developed a comprehensive search strategy to find relevant articles from electronic databases like PubMed, Google Scholar, Web of Science (WoS), or Scopus. We looked through the literature and determined which diagnostic methods in cancer genetics were particularly reliable. We used phrases like 'cancer genetics', genetic susceptibility, Hereditary cancer, cancer risk assessment, 'cancer diagnostic tools', cancer screening', biomarkers, and molecular diagnostics, reviews and meta-analyses evaluating the efficacy and safety of molecular therapies for cancer-related disorders. Research that only consider treatment modalities that don't necessitate genetic or molecular diagnostics fall under the exclusion criteria. RESULTS The results of this comprehensive review clearly demonstrate the transformative impact of molecular methods in the realm of cancer genetics.This review underscores how these technologies have empowered researchers and clinicians to identify and understand key genetic alterations that drive malignancy, ranging from point mutations to structural variations. Such insights are instrumental in pinpointing critical oncogenic drivers and potential therapeutic targets, thus opening the door for methods in precision medicine that can significantly improve patient outcomes. LIMITATION The search does not specify a timeframe for publication inclusion, it may have missed recent advancements or changes in the field's landscape of molecular methods for cancer. As a result, it may not have included the most recent developments in the field. CONCLUSION After conducting an in-depth study on the molecular methods in cancer genetics, it is evident that these cutting-edge technologies have revolutionized the field of oncology, providing researchers and clinicians with powerful tools to unravel the complexities of cancer at the genetic level. The integration of molecular methods techniques has not only enhanced our understanding of cancer etiology, progression, and treatment response but has also opened new avenues for personalized medicine and targeted therapies, leading to improved patient outcomes.
Collapse
Affiliation(s)
- Tanushree Budhbaware
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| | - Jaishriram Rathored
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| | - Sandesh Shende
- Department of ‘School of Allied Health Sciences’, Central Research Laboratory (CRL) and Molecular Diagnostics, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, India
| |
Collapse
|
174
|
Song M, Yoon H, Yoon H, Lee HM, Chae YJ, Chang JE. Enhanced anticancer efficacy of photodynamic therapy in combination with immunotherapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 261:113048. [PMID: 39476746 DOI: 10.1016/j.jphotobiol.2024.113048] [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: 08/29/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 12/10/2024]
Abstract
Photodynamic therapy (PDT) is known to trigger immunogenic cell death (ICD), leading to an anticancer effect even in untreated metastatic cancer, a phenomenon called the "abscopal effect". Furthermore, ICD induction activates an immune response, which may synergize with immunotherapy. The objective of our research was to evaluate the anticancer efficacy of combining PDT with immunotherapy. To assess in vivo anticancer efficacy and the abscopal effect, we implanted CT26 cells on both flanks of BALB/c mice. The mice were categorized into five different groups: 1) PBS, 2) immunotherapy alone, 3) PDT alone, 4) immunotherapy administered 3 days after PDT, and 5) immunotherapy administered immediately after PDT. The observed antitumor effects on the primary tumor followed this order: immunotherapy administered immediately after PDT > immunotherapy administered 3 days after PDT > PDT alone > immunotherapy alone > PBS. In metastatic tumors that were not directly treated, immunotherapy administered immediately after PDT was also the most effective. In conclusion, our study confirms that the combination of PDT with immunotherapy enhances anticancer efficacy against both primary and metastatic tumors. Additionally, administering immunotherapy immediately after PDT is more effective than delayed administration.
Collapse
Affiliation(s)
- Mihyun Song
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Heewon Yoon
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Hyejin Yoon
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Hyang-Mi Lee
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea
| | - Ji-Eun Chang
- College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea.
| |
Collapse
|
175
|
Kadian LK, Verma D, Lohani N, Yadav R, Ranga S, Gulshan G, Pal S, Kumari K, Chauhan SS. Long non-coding RNAs in cancer: multifaceted roles and potential targets for immunotherapy. Mol Cell Biochem 2024; 479:3229-3254. [PMID: 38413478 DOI: 10.1007/s11010-024-04933-1] [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: 06/06/2023] [Accepted: 01/05/2024] [Indexed: 02/29/2024]
Abstract
Cancer remains a major global health concern with high mortality rates mainly due to late diagnosis and poor prognosis. Long non-coding RNAs (lncRNAs) are emerging as key regulators of gene expression in human cancer, functioning through various mechanisms including as competing endogenous RNAs (ceRNAs) and indirectly regulating miRNA expression. LncRNAs have been found to have both oncogenic and tumor-suppressive roles in cancer, with the former promoting cancer cell proliferation, migration, invasion, and poor prognosis. Recent research has shown that lncRNAs are expressed in various immune cells and are involved in cancer cell immune escape and the modulation of the tumor microenvironment, thus highlighting their potential as targets for cancer immunotherapy. Targeting lncRNAs in cancer or immune cells could enhance the anti-tumor immune response and improve cancer immunotherapy outcomes. However, further research is required to fully understand the functional roles of lncRNAs in cancer and the immune system and their potential as targets for cancer immunotherapy. This review offers a comprehensive examination of the multifaceted roles of lncRNAs in human cancers, with a focus on their potential as targets for cancer immunotherapy. By exploring the intricate mechanisms underlying lncRNA-mediated regulation of cancer cell proliferation, invasion, and immune evasion, we provide insights into the diverse therapeutic applications of these molecules.
Collapse
Affiliation(s)
- Lokesh K Kadian
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
- Dept of Dermatology, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Deepika Verma
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neelam Lohani
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ritu Yadav
- Dept of Genetics, MD University, Rohtak, 124001, India
| | - Shalu Ranga
- Dept of Genetics, MD University, Rohtak, 124001, India
| | - Gulshan Gulshan
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Sanghapriya Pal
- Dept of Biochemistry, Maulana Azad Medical College and Associated Hospital, New Delhi, 110002, India
| | - Kiran Kumari
- Dept of Forensic Science, Lovely Professional University, Jalandhar, Punjab, 144411, India
| | - Shyam S Chauhan
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
176
|
Kassie BB, Getahun MJ, Azanaw A, Ferede BT, Tassew DF. Surface modification of cellulose nanocrystals for biomedical and personal hygiene applications. Int J Biol Macromol 2024; 282:136949. [PMID: 39490486 DOI: 10.1016/j.ijbiomac.2024.136949] [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: 09/01/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
The increasing demand for sustainable and effective materials in biomedical and personal hygiene applications has driven the exploration of cellulose nanocrystals (CNCs) derived from biomass. These nanomaterials are highly valued for their exceptional mechanical properties, biocompatibility, and renewable nature. Researchers are exploring CNCs for advancing medical and hygiene products, but surface modification is often needed to maximize their benefits. Techniques such as chemical functionalization, physical coating, and hybridization can significantly enhance CNCs dispersibility, stability, and interaction with biological systems. This versatility makes CNCs suitable for a variety of applications, including drug delivery systems, wound dressings, and personal hygiene products. Despite their advantages, maintaining the inherent properties of CNCs while integrating new functionalities through modification poses a challenge. Understanding the impact of various modification techniques on CNC performance is crucial for optimizing their effectiveness. This review aimed to consolidate current knowledge on the surface modification of biomass-derived CNCs, offering insights into different methods and their implications for biomedical and personal hygiene applications. By highlighting advancements, challenges, and prospects, it served as a crucial resource for advancing the development and application of CNCs in these critical fields.
Collapse
Affiliation(s)
- Bantamlak Birlie Kassie
- Textile Faculty, Ethiopian Institute of Textile and Fashion Technology, Bahir Dar University, Bahir Dar, P.O. Box 1037, Ethiopia; Medical Textile Research Center, Ethiopian Institute of Textile and Fashion Technology, Bahir Dar University, Bahir Dar, P.O. Box 1037, Ethiopia.
| | | | - Aklilu Azanaw
- Textile Faculty, Ethiopian Institute of Textile and Fashion Technology, Bahir Dar University, Bahir Dar, P.O. Box 1037, Ethiopia
| | - Bayu Teshome Ferede
- Textile Faculty, Ethiopian Institute of Textile and Fashion Technology, Bahir Dar University, Bahir Dar, P.O. Box 1037, Ethiopia
| | - Dehenenet Flatie Tassew
- Textile Faculty, Ethiopian Institute of Textile and Fashion Technology, Bahir Dar University, Bahir Dar, P.O. Box 1037, Ethiopia
| |
Collapse
|
177
|
Girardi FM, Pesenatto GG, Fuchs LD, Maahs TP, Treister NS, Maahs GS. Navigating Treatment‐Induced Head and Neck Malignancies in Cancer Survivors. Oral Dis 2024. [DOI: 10.1111/odi.15213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
ABSTRACTCancer presents a significant global health burden. Traditional treatment modalities, especially radiotherapy and chemotherapy, have improved survival, but they have long‐term consequences. Treatment‐induced malignancies, a growing concern, pose risks to survivors. Often with limited treatment options, these tumors tend to have a worse survival rate. A thorough understanding of the main risk groups is critical for lifelong follow‐up and personalized care strategies. This review provides an update on the knowledge of treatment‐induced malignancies of the head and neck in cancer survivors; specifically, it discusses the risks, histological considerations, diagnosis and treatment strategies, follow‐up recommendations, and future perspectives.
Collapse
Affiliation(s)
- Fábio Muradás Girardi
- Head and Neck Surgery Department Ana Nery Hospital Santa Cruz do Sul Rio Grande do Sul Brazil
| | - Gustavo Guthmann Pesenatto
- Postgraduate Program in Otorhinolaryngology Hospital São Lucas da PUCRS Porto Alegre Rio Grande do Sul Brazil
| | - Larissa Dill Fuchs
- School of Dentistry Universidade Federal do Rio Grande do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Thomas Peter Maahs
- Otorhinolaryngology and Head and Neck Surgery Department Pontifícia Universidade Católica Do Rio Grande Do Sul Porto Alegre Rio Grande do Sul Brazil
| | - Nathaniel S. Treister
- Division of Oral Medicine and Dentistry Brigham and Women's Hospital Boston Massachusetts USA
- Department of Oral Medicine, Infection and Immunity Harvard School of Dental Medicine Boston Massachusetts USA
| | - Gerson Schulz Maahs
- Otorhinolaryngology and Head and Neck Surgery Department Pontifícia Universidade Católica Do Rio Grande Do Sul Porto Alegre Rio Grande do Sul Brazil
- Otorhinolaryngology and Head and Neck Surgery Department Hospital de Clínicas de Porto Alegre Porto Alegre Rio Grande do Sul Brazil
| |
Collapse
|
178
|
Sai Madhurya M, Thakur V, Dastari S, Shankaraiah N. Pyrrolo[2,3-d]pyrimidines as potential kinase inhibitors in cancer drug discovery: A critical review. Bioorg Chem 2024; 153:107867. [PMID: 39388837 DOI: 10.1016/j.bioorg.2024.107867] [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: 07/03/2024] [Revised: 08/23/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Pyrrolo[2,3-d]pyrimidine-based kinase inhibitors have emerged as an important class of targeted therapeutics to combat various types of cancer. The distinctive structural feature of pyrrolopyrimidine ring system offers an adaptable platform for designing potent inhibitors of various kinases, crucial in regulating cellular processes. The deazapurine framework inherent to pyrrolopyrimidines bears a conspicuous resemblance to adenine, the natural ligand ATP. The structural mimicry enhances their appeal as potent inhibitors of key kinases. This review reconnoitres the intricate process of designing and developing pyrrolopyrimidine based derivatives, accentuating their structural diversity and the strategic modifications employed to enhance selectivity, potency, and pharmacokinetic properties. The discussion delves into medicinal chemistry strategies, highlighting successful examples that have been progressed to clinical evaluation. Furthermore, the review highlights the promise of pyrrolopyrimidine scaffolds in revolutionizing targeted cancer therapy and provides a pioneering perspective on future directions.
Collapse
Affiliation(s)
- Malyala Sai Madhurya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Vanashree Thakur
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Sowmya Dastari
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
179
|
Minh PH, Anh PTV, Tung BT, Dung HM, Trang TTT, Nhung PTH, Hang NT, Nguyet NTM, Phong NV, Vinh LB, Thanh MP. Efficacy of Jasminum subtriplinerve Extract against 7,12-Dimethylbenz[ a]anthracene-Induced Cancer in Mice. J Microbiol Biotechnol 2024; 34:2173-2183. [PMID: 39317690 PMCID: PMC11637824 DOI: 10.4014/jmb.2407.07045] [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: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
Jasminum subtriplinerve Blume tea is a traditional Vietnamese medicine used to treat impetigo, menstruation issues, and painful menstrual hematometra. Previous studies have shown that extracts and isolated compounds from J. subtriplinerve possess diverse pharmacological properties, such as antioxidant, antibacterial, and antidiabetic effects. However, their potential anticancer effects and underlying mechanisms of action have not been clear. Here, we examined the effects of J. subtriplinerve extracts against three human cancer cell lines. We also conducted in vivo analyses using a mouse model of 7,12-dimethylbenz[a]anthracene-induced breast cancer, including an investigation of changes in histological sections. The effect of the J. subtriplinerve ethyl acetate fraction on cytokine levels (IL-2, PGE2, TNF-α) in serum was determined using ELISA kits. Results showed that the ethyl acetate (EtOAc) fraction had the highest anti-proliferative activity (IC50 = 13.7 mg/ml) against the breast cancer (MCF-7) cell line, while the butanol (BuOH) and water fractions did not show any anticancer effects. Additionally, the EtOAc fraction at a dose of 14.4 mg/kg was able to elevate IL-2 levels and suppress the expression of PGE2 in the serum of mice. A remarkable decrease in the percentage of death and tumor incidence in mice was achieved following treatment with the EtOAc fraction at a dose of 14.4mg/kg. No abnormal parameters in blood were observed in the J. subtriplinerve treatment groups. These results suggest that J. subtriplinerve, when used as tea or a functional food, is nontoxic and has clear chemopreventive effects against breast cancer.
Collapse
Affiliation(s)
- Phan Hong Minh
- University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
- Department of Pharmacology, Hanoi Medical University, Hanoi, Vietnam
| | - Pham Thi Van Anh
- Department of Pharmacology, Hanoi Medical University, Hanoi, Vietnam
| | - Bui Thanh Tung
- University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Ho My Dung
- University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | | | - Pham Thi Hong Nhung
- University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Nguyen Thi Hang
- National Institute of Medical Materials (NIMM), Hanoi, 11022, Vietnam
| | | | - Nguyen Viet Phong
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Le Ba Vinh
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Mai Phuong Thanh
- Department of Pharmacology, Hanoi Medical University, Hanoi, Vietnam
| |
Collapse
|
180
|
Pathirana OC, Paranagama MP, Wijesundera KK, Mahakapuge TAN, Abeykoon AMAU, Rajapakse J. Elucidating the potential of Annona muricata L. grown in Sri Lanka to be used in developing an anticancer drug against colorectal and breast cancers. BMC Complement Med Ther 2024; 24:410. [PMID: 39609783 PMCID: PMC11603727 DOI: 10.1186/s12906-024-04712-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Since ancient times many traditional medicine systems around the world have been using different parts of Annona muricata L. (AM), to treat cancer. Indeed, numerous in vitro and in vivo studies also have shown anticancer properties of different solvent extracts of different parts of AM. Even the same part of the plant has shown different levels of anticancer properties based on geographical variations. Therefore, in the present study, the anticancer potential of the leaves, fruit pulp and the fruit peel of the AM that is grown in Sri Lanka was comparatively analyzed with the intention of identifying the most suitable part to be developed into a nutraceutical with anticancer effects. METHODS Freeze-dried aqueous extracts of immature leaves (ILAM), mature leaves (MLAM), pulp (PAM) and peel (PLAM) of AM were analyzed for their antioxidant activity using 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging and 2,2-azinobis-3-ethylbenzothiozoline-6-sulfonic acid (ABTS) cation decolorization assays. Their cytotoxicity on breast cancer (MCF-7) cells, colorectal adenocarcinoma (DLD-1) cells and normal human gingival fibroblasts (HGF-1) were determined by the 3-(4,5- dimethylthiazole-2-yl)-2,5-diphenyl-tetrazoliumbromide (MTT) assay. Their effect on mRNA expression of proapoptotic (Bax and caspase-7) and cell cycle arresting (p21) genes was analyzed by RT- qPCR in the same cell lines. RESULTS ILAM demonstrated the highest antioxidant activity in both DPPH and ABTS assays followed by MLAM, PLAM and PAM. In the MTT assay, both ILAM and MLAM demonstrated strong cytotoxic activity against MCF-7 and DLD-1 cell lines while there were no cytotoxic effects on the normal human cell line HGF-1. Both ILAM and MLAM demonstrated concentration-dependent upregulation of mRNA expression of cell cycle arresting gene p21 and apoptosis inducing genes Bax and caspase-7 in MCF-7 and DLD-1 cells. CONCLUSION The AEAM leaves grown in Sri Lanka has significantly higher antioxidant activity as well as selective cytotoxic effects on MCF-7 and DLD-1 cancer cells compared to its PL and P counterparts. Further, the AEAM leaves induced mRNA expression of the anticancer genes p21, Bax and caspase-7, indicating its potential to be developed into an anticancer drug against breast and colorectal cancer.
Collapse
Affiliation(s)
- Onela Canith Pathirana
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka.
| | - Madhavi Priyanka Paranagama
- Department of Basic Sciences, Faculty of Dental Sciences, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Kavindra Kumara Wijesundera
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | | | - Jayantha Rajapakse
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| |
Collapse
|
181
|
Padnya P, Shiabiev I, Pysin D, Gerasimova T, Ranishenka B, Stanavaya A, Abashkin V, Shcharbin D, Shi X, Shen M, Nazarova A, Stoikov I. Non-Viral Systems Based on PAMAM-Calix-Dendrimers for Regulatory siRNA Delivery into Cancer Cells. Int J Mol Sci 2024; 25:12614. [PMID: 39684325 DOI: 10.3390/ijms252312614] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer is one of the most common diseases in developed countries. Recently, gene therapy has emerged as a promising approach to cancer treatment and has already entered clinical practice worldwide. RNA interference-based therapy is a promising method for cancer treatment. However, there are a number of limitations that require vectors to deliver therapeutic nucleic acids to target tissues and organs. Active research is currently underway to find highly effective, low-toxic nanomaterials capable of acting as nanocarriers. In this study, we demonstrated for the first time the ability of symmetrical polyamidoamine dendronized thiacalix[4]arenes (PAMAM-calix-dendrimers) to form stable positively charged complexes with siRNAs, protect them from enzymatic degradation, and efficiently deliver gene material to HeLa cells. A distinctive feature of PAMAM-calix-dendrimers was the unusual decrease in hemo- and cytotoxicity with increasing generation, while these compounds did not cause toxic effects at concentrations required for siRNA binding and delivery. A comparative analysis of the efficiency of complex formation of PAMAM-calix-dendrimers and classical PAMAM dendrimers with siRNAs was also performed. The findings may facilitate the creation of novel unique gene delivery systems for cancer nanomedicine development.
Collapse
Affiliation(s)
- Pavel Padnya
- A.M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Igor Shiabiev
- A.M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Dmitry Pysin
- A.M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Tatiana Gerasimova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, 420088 Kazan, Russia
| | - Bahdan Ranishenka
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya St., 220072 Minsk, Belarus
| | - Alesia Stanavaya
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya St., 220072 Minsk, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya St., 220072 Minsk, Belarus
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya St., 220072 Minsk, Belarus
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- CQM-Centro de Química da Madeira, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Anastasia Nazarova
- A.M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| | - Ivan Stoikov
- A.M. Butlerov Chemistry Institute, Kazan Federal University, 18 Kremlyovskaya Str., 420008 Kazan, Russia
| |
Collapse
|
182
|
Oliveira I, Rodrigues-Santos P, Ferreira L, Pires das Neves R. Synthetic and biological nanoparticles for cancer immunotherapy. Biomater Sci 2024; 12:5933-5960. [PMID: 39441658 DOI: 10.1039/d4bm00995a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Cancer is becoming the main public health problem globally. Conventional chemotherapy approaches are slowly being replaced or complemented by new therapies that avoid the loss of healthy tissue, limit off-targets, and eradicate cancer cells. Immunotherapy is nowadays an important strategy for cancer treatment, that uses the host's anti-tumor response by activating the immune system and increasing the effector cell number, while, minimizing cancer's immune-suppressor mechanisms. Its efficacy is still limited by poor therapeutic targeting, low immunogenicity, antigen presentation deficiency, impaired T-cell trafficking and infiltration, heterogeneous microenvironment, multiple immune checkpoints and unwanted side effects, which could benefit from improved delivery systems, able to release immunotherapeutic agents to tumor microenvironment and immune cells. Nanoparticles (NPs) for immunotherapy (Nano-IT), have a huge potential to solve these limitations. Natural and/or synthetic, targeted and/or stimuli-responsive nanoparticles can be used to deliver immunotherapeutic agents in their native conformations to the site of interest to enhance their antitumor activity. They can also be used as co-adjuvants that enhance the activity of IT effector cells. These nanoparticles can be engineered in the natural context of cell-derived extracellular vesicles (EVs) or exosomes or can be fully synthetic. In this review, a detailed SWOT analysis is done through the comparison of engineered-synthetic and naturaly-derived nanoparticles in terms of their current and future use in cancer immunotherapy.
Collapse
Affiliation(s)
- Inês Oliveira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Paulo Rodrigues-Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
183
|
Nujoom N, Koyakutty M, Biswas L, Rajkumar T, Nair SV. Emerging Gene-editing nano-therapeutics for Cancer. Heliyon 2024; 10:e39323. [PMID: 39524822 PMCID: PMC11550658 DOI: 10.1016/j.heliyon.2024.e39323] [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: 12/08/2023] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Remarkable progress has been made in the field of genome engineering after the discovery of CRISPR/Cas9 in 2012 by Jennifer Doudna and Emmanuelle Charpentier. Compared to any other gene-editing tools, CRISPR/Cas9 attracted the attention of the scientific community because of its simplicity, specificity, and multiplex editing possibilities for which the inventors were awarded the Nobel prize for chemistry in 2020. CRISPR/Cas9 allows targeted alteration of the genomic sequence, gene regulation, and epigenetic modifications using an RNA-guided site-specific endonuclease. Though the impact of CRISPR/Cas9 was undisputed, some of its limitations led to key modifications including the use of miniature-Cas proteins, Cas9 Retron precise Parallel Editing via homologY (CRISPEY), Cas-Clover, or development of alternative methods including retron-recombineering, Obligate Mobile Element Guided Activity(OMEGA), Fanzor, and Argonaute proteins. As cancer is caused by genetic and epigenetic alterations, gene-editing was found to be highly useful for knocking out oncogenes, editing mutations to regain the normal functioning of tumor suppressor genes, knock-out immune checkpoint blockade in CAR-T cells, producing 'off-the-shelf' CAR-T cells, identify novel tumorigenic genes and functional analysis of multiple pathways in cancer, etc. Advancements in nanoparticle-based delivery of guide-RNA and Cas9 complex to the human body further enhanced the potential of CRISPR/Cas9 for clinical translation. Several studies are reported for developing novel delivery methods to enhance the tumor-specific application of CRISPR/Cas9 for anticancer therapy. In this review, we discuss new developments in novel gene editing techniques and recent progress in nanoparticle-based CRISPR/Cas9 delivery specific to cancer applications.
Collapse
Affiliation(s)
- Najma Nujoom
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Manzoor Koyakutty
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Lalitha Biswas
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Thangarajan Rajkumar
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| | - Shantikumar V. Nair
- Amrita School of Nanosciences and Molecular Medicine, Amrita Vishwavidyapeetham (University), Ponekkara P.O., Kochi, India
| |
Collapse
|
184
|
Chandra J, Nasir N, Wahab S, Sahebkar A, Kesharwani P. Harnessing the power of targeted metal nanocarriers mediated photodynamic and photothermal therapy. Nanomedicine (Lond) 2024:1-19. [PMID: 39545609 DOI: 10.1080/17435889.2024.2419820] [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: 04/25/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
The treatment of cancer has become a profoundly intricate procedure. Traditional treatment methods, including chemotherapy, surgery and radiotherapy, have been utilized, while notable progress has been achieved in recent years. Among targeted therapies for cancer, folic acid (FA) conjugated metal-based nanoparticles (NP) have emerged as an innovative strategy, namely for photodynamic therapy (PDT) and photothermal therapy (PTT). These NP exploit the strong attraction between FA and folate receptors, which are excessively produced in several cancer cells, in order to enable precise administration and improved effectiveness of treatment. During PDT, metal-based NP functionalized with FA are used as photosensitizers which are activated by light, and produce reactive oxygen species that cause cancer cells to undergo apoptosis. Within the framework of PTT, these NP effectively transform light energy into concentrated heat, specifically targeting and destroying tumor cells. This review examines the fundamental mechanisms by which these NP improve the effectiveness of PDT and PTT while simultaneously presenting important findings that demonstrate the effectiveness of FA-functionalized MNP in laboratory and animal models. In addition, the paper also discusses the problems and potential directions for their clinical translation.
Collapse
Affiliation(s)
- Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazim Nasir
- Department of Basic Medical Sciences, College of Applied Medical Sciences, Khamis Mushait, King Khalid University, Abha, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 61421, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
185
|
Gharatape A, Amanzadi B, Mohamadi F, Rafieian M, Faridi-Majidi R. Recent advances in polymeric and lipid stimuli-responsive nanocarriers for cell-based cancer immunotherapy. Nanomedicine (Lond) 2024; 19:2655-2678. [PMID: 39540464 DOI: 10.1080/17435889.2024.2416377] [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: 08/09/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Conventional cancer therapy has major limitations, including non-specificity, unavoidable side effects, low specific tumor accumulation and systemic toxicity. In recent years, more effective and precise treatment methods have been developed, including cell-based immunotherapy. Carriers that can accurately and specifically target cells and equip them to combat cancer cells are particularly important for developing this therapy. As a result, attention has been drawn to smart nanocarriers that can react to specific stimuli. Thus, stimuli-responsive nanocarriers have attracted increasing attention because they can change their physicochemical properties in response to stimulus conditions, such as pH, enzymes, redox agents, hypoxia, light and temperature. This review highlights recent advances in various stimuli-responsive nanocarriers, discussing loading, targeted delivery, cellular uptake, biocompatibility and immunomodulation in cell-based immunotherapy. Finally, future challenges and perspectives regarding the possible clinical translation of nanocarriers are discussed.
Collapse
Affiliation(s)
- Alireza Gharatape
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Bentolhoda Amanzadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Faranak Mohamadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Mahdieh Rafieian
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Reza Faridi-Majidi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Pharmaceutical Nanotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
186
|
Wang Z, Yue S, Chen X, Li J, Zhu P, Chen H, Qiu F, Xie D, Liang Y, Li D, Lu A, Liang C. Design of Murine Double Minute 2 Proteolysis Targeting Chimera Degraders with a Built-In Tumor-Targeting Ability. J Med Chem 2024; 67:18865-18882. [PMID: 39437434 DOI: 10.1021/acs.jmedchem.4c01228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional molecules to induce the proteasomal degradation of target proteins. Currently, there are no tumor-targeting PROTACs for modulating oncogenic murine double minute 2 (MDM2). AS1411 is a tumor-targeting aptamer that specifically recognizes nucleolin (NCL) overexpressed on the surface of tumor cells. We recently repurposed AS1411 as an MDM2 recruiter since it could form an NCL-bridged ternary complex with MDM2. In this study, we design a PROTAC molecule AS1411-VH032 via conjugating AS1411 with a recruiter of von Hippel-Lindau (VHL) ligase VH032. AS1411-VH032 facilitates tumor-selective degradation of MDM2, leading to tumor shrinkage with no detectable toxicity. Besides being a molecular target, MDM2 also serves as an E3 ligase harnessed by PROTACs. Thus, we developed an AS1411-based homo-PROTAC homoAS1411, which induces tumor-specific suicide degradation of MDM2 and prevents tumor progression without causing side effects. Both AS1411-VH032 and homoAS1411 are promising MDM2 degraders with built-in tumor-targeting ability, which balances the antitumor efficacy with a favorable safety profile.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jin Li
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Peixi Zhu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yiying Liang
- Shenzhen LingGene Biotech Co., Ltd., Shenzhen 518055, China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| |
Collapse
|
187
|
Ebrahim NAA, Abou-Bakr AA, Tawfik HN, Nassar HR, Adel I. Decoding β-catenin expression patterns in ovarian serous carcinoma with clinicopathological implications: insights from National Cancer Institute. Clin Transl Oncol 2024:10.1007/s12094-024-03770-4. [PMID: 39538105 DOI: 10.1007/s12094-024-03770-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
AIM This study aimed to examine the immunohistochemical expression of β-catenin in serous ovarian carcinoma and to investigate its relationship with clinicopathological features and disease outcomes. METHODS A retrospective analysis was conducted on 67 cases of serous ovarian carcinoma diagnosed at the Pathology Department of the Egyptian National Cancer Institute, Cairo University, between January 1, 2015, and December 31, 2017. RESULTS The age of the patients ranged from 26 to 76 years. Aberrant β-catenin expression was defined by the presence of cytoplasmic staining with or without membranous staining in at least 10% of tumor cells. Of the cases analyzed, thirty exhibited cytoplasmic staining, 18 demonstrated preserved expression on the cell membrane, 15 showed combined cytoplasmic and membranous staining, while four cases were entirely negative. Significant correlations were observed between β-catenin positivity and both tumor grade and p53 expression, with p values of 0.004 for each correlation. CONCLUSIONS Positive β-catenin expression significantly correlated with tumor grade in serous ovarian carcinoma. Most low-grade serous carcinoma cases exhibited membranous β-catenin expression, whereas high-grade serous carcinoma cases predominantly displayed cytoplasmic staining. Therapeutic strategies aimed at inhibiting β-catenin signaling could provide a novel approach to improving outcomes for patients with high-grade ovarian cancer.
Collapse
Affiliation(s)
- Noura A A Ebrahim
- Oncologic Pathology Department, National Cancer Institute (NCI), Cairo University, Cairo, Egypt.
| | - Amany A Abou-Bakr
- Oncologic Pathology Department, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Hassan N Tawfik
- Oncologic Pathology Department, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Hanan R Nassar
- Medical Oncology Department, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Iman Adel
- Oncologic Pathology Department, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| |
Collapse
|
188
|
Li D, Hu S, Ye J, Zhai C, Liu J, Wang Z, Zhou X, Chen L, Zhou F. The Emerging Role of IGF2BP2 in Cancer Therapy Resistance: From Molecular Mechanism to Future Potential. Int J Mol Sci 2024; 25:12150. [PMID: 39596216 PMCID: PMC11595103 DOI: 10.3390/ijms252212150] [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: 10/17/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Tumor resistance is one of the primary reasons for cancer treatment failure, significantly limiting the options and efficacy of cancer therapies. Therefore, overcoming resistance has become a critical factor in improving cancer treatment outcomes. IGF2BP2, as a reader of m6A methylation, plays a pivotal role in the post-transcriptional regulation of RNA through the methylation of m6A sites. It not only contributes to cancer initiation and progression but also plays a key role in tumor drug resistance. This review provides a comprehensive summary of the mechanisms by which IGF2BP2 contributes to therapy resistance, with the aim of improving the efficacy of chemotherapy in cancer treatment. Advancing research in this area is crucial for developing more effective therapies that could significantly improve the quality of life for cancer patients.
Collapse
Affiliation(s)
- Die Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Shiqi Hu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200433, China
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Chaojie Zhai
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Medical Center for Cardiovascular Diseases, Neurological Diseases and Tumors of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Fan Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; (D.L.); (S.H.); (J.Y.); (C.Z.); (J.L.); (Z.W.); (X.Z.)
| |
Collapse
|
189
|
Nazari A, Osati P, Seifollahy Fakhr S, Faghihkhorasani F, Ghanaatian M, Faghihkhorasani F, Rezaei-Tazangi F, Pazhouhesh Far N, Shourideh A, Ebrahimi N, Aref AR. New Emerging Therapeutic Strategies Based on Manipulation of the Redox Regulation Against Therapy Resistance in Cancer. Antioxid Redox Signal 2024. [PMID: 39506926 DOI: 10.1089/ars.2023.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Background: Resistance to standard therapeutic methods, including chemotherapy, immunotherapy, and targeted therapy, remains a critical challenge in effective cancer treatment. Redox homeostasis modification has emerged as a promising approach to address medication resistance. Objective: This review aims to explore the mechanisms of redox alterations and signaling pathways contributing to treatment resistance in cancer. Methods: In this study, a comprehensive review of the molecular mechanisms underlying drug resistance governed by redox signaling was conducted. Emphasis was placed on understanding how tumor cells manage increased reactive oxygen species (ROS) levels through upregulated antioxidant systems, enabling resistance across multiple therapeutic pathways. Results: Key mechanisms identified include alterations in drug efflux, target modifications, metabolic changes, enhanced DNA damage repair, stemness preservation, and tumor microenvironment remodeling. These pathways collectively facilitate tumor cells' adaptive response and resistance to various cancer treatments. Conclusion: Developing a detailed understanding of the interrelationships between these redox-regulated mechanisms and therapeutic resistance holds potential to improve treatment effectiveness, offering valuable insights for both fundamental and clinical cancer research. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Parisa Osati
- Department of Chemical Engineering, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Norway
| | - Ferdos Faghihkhorasani
- Department of Cardiology, Internal Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, 710061, China
| | - Masoud Ghanaatian
- Master 1 Bio-Santé-Parcours Toulouse Graduate School of Cancer, Ageing and Rejuvenation (CARe), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Fereshteh Faghihkhorasani
- General Physician in Medicine Program,General Doctorate Degree of Yazd Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Amir Shourideh
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA and Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
190
|
Al-Azzawi HMA, Hamza SA, Paolini R, Arshad F, Patini R, O'Reilly L, McCullough M, Celentano A. Towards an emerging role for anticoagulants in cancer therapy: a systematic review and meta-analysis. FRONTIERS IN ORAL HEALTH 2024; 5:1495942. [PMID: 39568788 PMCID: PMC11576436 DOI: 10.3389/froh.2024.1495942] [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: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024] Open
Abstract
Background Anticoagulants, renowned for their role in preventing blood clot formation, have captivated researchers' attention for the exploitation of their potential to inhibit cancer in pre-clinical models. Objectives To undertake a systematic review and meta-analysis of the effects of anticoagulants in murine cancer research models. Further, to present a reference tool for anticoagulant therapeutic modalities relating to future animal pre-clinical models of cancer and their translation into the clinic. Methods Four databases were utilized including Medline (Ovid), Embase (Ovid), Web of science, and Scopus databases. We included studies relating to any cancer conducted in murine models that assessed the effect of traditional anticoagulants (heparin and its derivatives and warfarin) and newer oral anticoagulants on cancer. Results A total of 6,158 articles were identified in an initial multi-database search. A total of 157 records were finally included for data extraction. Studies on heparin species and warfarin demonstrated statistically significant results in favour of tumour growth and metastasis inhibition. Conclusion Our findings constitute a valuable reference guide for the application of anticoagulants in cancer research and explore the promising utilization of non-anticoagulants heparin in preclinical cancer research. Systematic Review Registration PROSPERO [CRD42024555603].
Collapse
Affiliation(s)
| | - Syed Ameer Hamza
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Fizza Arshad
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Romeo Patini
- Head and Neck Department, "Fondazione Policlinico Universitario A. Gemelli-IRCCS" School of Dentistry, Catholic University of Sacred Heart-Rome Largo A. Gemelli, Rome, Italy
| | - Lorraine O'Reilly
- Clinical Translation Centre, Cancer Biology and Stem Cells Division and Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
191
|
Alqahtani MM. Cisplatin-Induced Renal Failure Measured by Glomerular Filtration Rate (GFR) with 99mTc-DTPA Scans in Cancer Patients: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2024; 14:2468. [PMID: 39594134 PMCID: PMC11593308 DOI: 10.3390/diagnostics14222468] [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: 09/03/2024] [Revised: 10/27/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Cisplatin is a potent agent commonly used to treat cancer, but its effects pose a significant risk to renal function. Therefore, the present study aimed to evaluate the impact of cisplatin on renal function as measured by glomerular filtration rate (GFR) using diethyltriamine-penta-acetic acid (DTPA) renal scintigraphy. Methods: Extensive literature searches were performed using PRISMA guidelines that investigated cisplatin-induced renal failure by measuring GFR with DTPA. Eligible studies were included based on predefined criteria. Data on GFR, serum creatinine levels, and acute kidney injury (AKI) before and after cisplatin therapy were extracted and analyzed. A meta-analysis was performed utilizing RevMan 5.4 to determine the overall effect of cisplatin on GFR before and after treatment. For non-randomized controlled trials (RCTs), quality assessment was performed using the Newcastle-Ottawa Scale, while for RCT, the Cochrane risk of bias tool was utilized. Results: Initially, 1003 studies were searched from different databases, including ScienceDirect, PubMed, Scopus, Google Scholar, and The Cochrane Library, and after screening, 8 studies (PubMed, Scopus, and GoogleS cholar) with 489 patients were found eligible for inclusion in the present study. Cisplatin was administrated with varying doses ranging from 20 mg/m2 to 114.02 mg/m2. The findings underscore the nephrotoxic effects of cisplatin, a widely used chemotherapeutic agent, as demonstrated by the significant decline in GFR observed across multiple treatment cycles, and these findings were also supported by the findings of a meta-analysis that showed a significant (p < 0.01) difference between peri- and post-treatment GFR level with 37.06 (95% CI, 10.90-63.23) effect size and 96% heterogeneity. In addition, the included studies were found to be of high quality. Conclusions: Cisplatin significantly affects renal function, as evidenced by a decrease in GFR measured with DTPA. The findings underscore the importance of the routine monitoring of GFR to detect early renal injury and guide treatment modification. Future research should focus on strategies to reduce cisplatin-induced toxicity and explore alternative therapies with reduced renal risk.
Collapse
Affiliation(s)
- Mansour M Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| |
Collapse
|
192
|
Barbosa-Azevedo M, Dias-Carvalho A, Carvalho F, Costa VM. Chemotherapy-induced cognitive impairment and glia: A new take on chemobrain? Toxicol Appl Pharmacol 2024; 492:117085. [PMID: 39236990 DOI: 10.1016/j.taap.2024.117085] [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: 02/23/2024] [Revised: 07/03/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
The significant rise in cancer survivorship stands out as one of the most notable achievements of modern science. However, this comes with a significant burden, as cancer treatment is not without adverse effects. Lately, there has been a growing focus on cognitive dysfunction associated with cancer treatment, often referred to as 'chemobrain'. It significantly impacts the quality of life for cancer survivors. The underlying mechanisms studied so far usually focus on neurons, while other cells of the central nervous system are often overlooked. This review seeks to place the hypothesis that glial cells may play a role in the development of chemotherapy-induced cognitive dysfunction. It summarizes the primary mechanisms proposed to date while underscoring the existing gaps in this research field. Inflammation and release of pro-inflammatory mediators by M1 microglia and A1 astrocytes are the most prevalent findings after chemotherapy. However, activation of A1 astrocytes by some chemotherapeutic agents may contribute to neuronal degeneration, alterations in synaptic branches, as well as glutamate excitotoxicity, which can contribute to cognitive impairment. Furthermore, the reduction in the number of oligodendrocytes after chemotherapy may also impact the myelin sheath, contributing to 'chemobrain'. Furthermore, some chemotherapeutic drugs activate M1 microglia, which is associated with decreased neuroplasticity and, possibly, cognitive impairment. In conclusion, data regarding the effects of chemotherapy on glial cells are scarce, and it is essential to understand how these cells are affected after cancer treatment to enable reliable therapeutic or preventive actions on cancer-treated patients.
Collapse
Affiliation(s)
- Maria Barbosa-Azevedo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
193
|
Qian J, Duan J, Cao D. Identification of a Novel 4-gene Prognostic Model Related to Neutrophil Extracellular Traps for Colorectal Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:849-858. [PMID: 39549020 PMCID: PMC11562497 DOI: 10.5152/tjg.2024.24131] [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: 03/14/2024] [Accepted: 06/25/2024] [Indexed: 11/18/2024]
Abstract
Background/Aims Colorectal cancer (CRC) is a significant global health concern, and understanding the molecular mechanisms underlying CRC progression and prognosis is crucial. Neutrophil extracellular traps (NETs) have been implicated in various cancers, but their role in CRC and its clinical implications remain to be elucidated. Materials and Methods Transcriptomic data from TCGA of CRC patients were analyzed to assess NETs enrichment and "NETs formation" pathway scores in NETs_high and NETs_low groups. Univariate Cox regression was used to identify prognosis-associated genes with the Log-Rank test for selection. Patients in the TCGA database were randomly split into training and testing sets to build a prognostic model with LASSO Cox regression. Model diagnostic performance was evaluated using Kaplan-Meier curves and receiver operating characteristic analysis. Single-sample gene set enrichment analysis (ssGSEA) was used to determine the abundance of 23 immune cells. ESTIMATE was used to calculate ImmuneScore and ESTIMATEScore, characterizing immune features of CRC samples. Results The NETs_high group in CRC showed significantly better survival than the NETs_low group. A robust prognostic model based on PRKRIP1, SERTAD2, ELFN1, and LINC00672 accurately predicted patient outcomes. NETs_high samples exhibited a more enriched immune environment with higher immune cell infiltration levels, as well as ImmuneScore and ESTIMATEScore. PRKRIP1, SERTAD2, ELFN1, and LINC00672 were significantly correlated with key immune cell types. Additionally, 18 drugs displayed differential sensitivity between NETs_high and NETs_low groups, with Daporinad and Selumetinib as potential therapeutic options. Conclusion Our findings may catalyze the development of personalized treatment modalities and bestow invaluable insights into the intricate dynamics governing CRC progression.
Collapse
Affiliation(s)
- Junwen Qian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, China
| | - Jiyun Duan
- Department of Breast Thyroid Head and Neck Surgery, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, China
| | - Dong Cao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, China
| |
Collapse
|
194
|
Hamdy NM, Basalious EB, El-Sisi MG, Nasr M, Kabel AM, Nossier ES, Abadi AH. Advancements in current one-size-fits-all therapies compared to future treatment innovations for better improved chemotherapeutic outcomes: a step-toward personalized medicine. Curr Med Res Opin 2024; 40:1943-1961. [PMID: 39412377 DOI: 10.1080/03007995.2024.2416985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/22/2024]
Abstract
The development of therapies followed a generalized approach for a long time, assuming that a single treatment could effectively address various patient populations. However, recent breakthroughs have revealed the limitations of this one-size-fits-all paradigm. More recently, the field of therapeutics has witnessed a shift toward other modules, including cell therapies, high molecular weight remedies, personalized medicines, and gene therapies. Such advancements in therapeutic modules have the potential to revolutionize healthcare and pave the way for medicines that are more efficient and with minimal side effects. Cell therapies have gained considerable attention in regenerative medicine. Stem cell-based therapies, for instance, hold promise for tissue repair and regeneration, with ongoing research focusing on enhancing their efficacy and safety. High molecular weight drugs like peptides and proteins emerged as promising therapeutics because of their high specificity and diverse biological functions. Engineered peptides and proteins are developed for targeted drug delivery, immunotherapy, and disease-modulation. In personalized medicine, tailored treatments to individuals based on specific genetic profiling, lifestyle, biomarkers, and disease characteristics are all implemented. Clinicians have tailored treatments to optimize outcomes and minimize adverse effects, using targeted therapies based on specific mutations, yielding remarkable results. Gene therapies have revolutionized the treatment of genetic disorders by directly targeting the underlying genetic abnormalities. Innovative techniques, such as CRISPR-Cas9 have allowed precise gene editing, opening up possibilities for curing previously incurable conditions. In conclusion, advancements in therapeutic modules have the potential to revolutionize healthcare and pave the way for medicines that are more efficient and with minimal side effects.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo, Egypt
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
| | - Emad B Basalious
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mona G El-Sisi
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo, Egypt
| | - Maha Nasr
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed M Kabel
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman S Nossier
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Ashraf H Abadi
- The National Committee of Drugs & Medicines by Academy of Scientific Research and Technology (ASRT), Ministry of Higher Education, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| |
Collapse
|
195
|
Mohamed DH, Said RS, Kassem DH, Gad AM, El-Demerdash E, Mantawy EM. Hesperidin attenuates radiation-induced ovarian failure in rats: Emphasis on TLR-4/NF-ĸB signaling pathway. Toxicol Appl Pharmacol 2024; 492:117111. [PMID: 39326792 DOI: 10.1016/j.taap.2024.117111] [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: 06/08/2024] [Revised: 09/09/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Young women suffering from premature ovarian failure after radiotherapy carry a huge burden in the field of cancer therapy including reproductive loss, emotional stress, and physical troubles that reduce their long-term quality of life. Hesperidin (HSP) exhibited antioxidant, anti-inflammatory, and anti-apoptotic properties. HSP enhanced in vitro follicular maturation and preserved in vivo ovarian stockpile. In this research, the role of HSP in radiation-induced POF in rats was investigated besides ascertaining its underlying mechanisms. Female Sprague-Dawley rats were arbitrarily allocated into four groups: control-group, ϒ-irradiated-group (3.2 Gy once on the 7th day), HSP-group (100 mg/kg, orally for 10 days), and HSP/ϒ-irradiated-group (ϒ-radiation was applied one hour after HSP). At the end of experiment, the whole ovaries were collected for histological and biochemical analyses. Administration of HSP preserved the ovarian histoarchitecture and follicular stock, retained ovarian weight, and conserved serum estradiol and AMH levels following radiation exposure. HSP ameliorated the ovarian oxidative damage mediated by radiation through augmenting the activities of glutathione peroxidase, glutathione reductase, and catalase antioxidant enzymes. HSP exhibited remarkable anti-inflammatory activity by downregulating the expression of ovarian TLR-4, NF-ĸB, and TNF-α. Moreover, HSP suppressed the apoptotic machinery triggered by radiation by reducing p53 and Bax while increasing Bcl-2 mRNA expressions alongside diminishing caspase-3 expression. Additionally, HSP regulated estrous cycle disorder of irradiated rats and improved their reproductive capacity reflected by enhancing pregnancy outcomes. Therefore, HSP represents an appealing candidate as an adjunct remedy for female cancer patients during radiotherapy protocols owing to its antioxidant, anti-inflammatory, anti-apoptotic, and hormone-regulatory effects.
Collapse
Affiliation(s)
- Doaa H Mohamed
- Central Administration of Drug Control, Egyptian Drug Authority, EDA, Formerly NODCAR, Giza, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Dina H Kassem
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amany M Gad
- Department of Pharmacology, Egyptian Drug Authority, EDA, Formerly NODCAR, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, Kantara Branch, Ismailia, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
196
|
Li Y, Yang X, Han T, Zhou J, Liu Y, Guo J, Liu Z, Bai Y, Xing Y, Ding X, Wu J, Hu D. IGFBP1 promotes the proliferation and migration of lung adenocarcinoma cells through the PPARα pathway. Transl Oncol 2024; 49:102095. [PMID: 39167955 PMCID: PMC11382126 DOI: 10.1016/j.tranon.2024.102095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND The immune status is closely linked to cancer progression, metastasis, and prognosis. Lipid metabolism, crucial for reshaping immune status, plays a key role in regulating the advancement of lung adenocarcinoma (LUAD) and deserves further investigation. METHODS This study classifies LUAD patients into different immune subtypes based on lipid metabolism-related genes and compares the clinical characteristics among these subtypes. Single-multi COX analysis screens out key genes related to prognosis, and a risk feature and prognostic model are constructed. Cell cloning, scratch, transwell, western blotting and flow cytometry cell cycle analysis to detect the function of key genes. A subcutaneous tumor animal model is used to investigate the in vivo function and molecular mechanisms of key genes. RESULTS LUAD patients are classified into three immune subtypes, among which C3 subtype has lower immune status and higher frequency of gene mutations, and show lower immunoreactivity in immunotherapy. COX analysis identified a prognostic model for four lipid metabolism factors (IGFBP1, NR0B2, PPARA, and POMC). IGFBP1, a core gene in this model, is highly expressed in the C3 subtype. Functionally, knocking down IGFBP1 significantly inhibits tumor cell cloning, scratch, and migration abilities, and downregulates the expression of cell cycle and EMT-related proteins. Knocking down IGFBP1 significantly inhibits tumor burden (P < 0.05). Mechanistically, knocking down IGFBP1 inhibits the activation of PPARα to regulate tumor cell growth. CONCLUSIONS This study found that lipid metabolism genes are closely related to LUAD, and IGFBP1 may be a key gene in regulating tumor growth and development.
Collapse
Affiliation(s)
- Yunyun Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Xuelian Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Ziqin Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China
| | - Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, China
| | - Yingru Xing
- Department of Clinical Laboratory, Anhui Zhongke Gengjiu Hospital, Hefei, China
| | - Xuansheng Ding
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; School of pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China.
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, China; Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, China; Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, China; Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, Anhui University of Science and Technology, Huainan, Anhui, China; Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, China.
| |
Collapse
|
197
|
Goleij P, Sanaye PM, Alam W, Zhang J, Tabari MAK, Filosa R, Jeandet P, Cheang WS, Efferth T, Khan H. Unlocking daidzein's healing power: Present applications and future possibilities in phytomedicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155949. [PMID: 39217652 DOI: 10.1016/j.phymed.2024.155949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cancer is one of the leading causes of death and a great threat to people around the world. Cancer treatment modalities include surgery, radiotherapy, chemotherapy, radiochemotherapy, hormone therapy, and immunotherapy. The best approach is to use a combination of several types. Among the treatment methods mentioned above, chemotherapy is frequently used, but its activity is hampered by the development of drug resistance and many side effects. In this regard, the use of medicinal plants has been discussed, and in recent decades, the use of isolated phytochemicals came into the focus of interest. By critically evaluating the available evidence and emphasizing the unique perspective offered by this review, we provide insights into the potential of daidzein as a promising therapeutic agent, as well as outline future research directions to optimize its efficacy in clinical settings. PURPOSE To summarized the therapeutic potential of daidzein, an isoflavone phytoestrogen in the management of several human diseases with the focuses on the current status and future prospects as a therapeutic agent. METHODS Several search engines, including PubMed, GoogleScholar, and ScienceDirect, were used, with the search terms "daidzein", "daidzein therapeutic potential", or individual effects. The study included all peer-reviewed articles. However, the most recent publications were given priority. RESULTS Daidzein showed protective effects against malignant diseases such as breast cancer, prostate cancer but also non-malignant diseases such as diabetes, osteoporosis, and cardiovascular diseases. Daidzein activates multiple signaling pathways leading to cell cycle arrest and apoptosis as well as antioxidant and anti-metastatic effects in malignant cells. Moreover, the anticancer effects against different cancer cells were more prominent and discussed in detail. CONCLUSIONS In short, daidzein represents a promising compound for drug development. The comprehensive potential anticancer activities of daidzein through various molecular mechanisms and its therapeutic/clinical status required further detail studies.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran.
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Waqas Alam
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Mohammad Amin Khazeei Tabari
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran; Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Rosanna Filosa
- Department of Science and Technology, University of Sannio, Benevento 82100, Italy
| | - Philippe Jeandet
- Département de Biologie et Biochimie Faculté des Sciences Exactes et Naturelles Université de Reims BP 1039 51687, Reims CEDEX 02, France
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| |
Collapse
|
198
|
Y KN, Arjunan A, Maigandan D, Dharmarajan A, Perumalsamy LR. Advances and challenges in therapeutic resistant biomarkers of neuroblastoma: A comprehensive review. Biochim Biophys Acta Rev Cancer 2024; 1879:189222. [PMID: 39577750 DOI: 10.1016/j.bbcan.2024.189222] [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: 06/06/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Therapeutic resistance is one of the significant challenges in Neuroblastoma. Owing to its molecular diversity, the therapeutic resistance mechanisms of Neuroblastoma are highly complicated. The traditional chemo and radio therapeutics fail to provide adequate solutions to the treatment resistance, demanding in-depth research to improvise the existing prognostic and therapeutic regimens. To address this knowledge gap, several investigations are being employed, such as unravelling the molecular signalling mechanisms involved in drug resistance at genomics and proteomics levels, development of biomarkers for assessing the therapeutic success, development of novel drug targets for cancer stem cells, targeted immunotherapy and combination therapies. This review collates the ongoing research efforts to address the challenges faced in Neuroblastoma treatment resistance and uncovers the importance of transitioning biomarker discoveries into clinical practice.
Collapse
Affiliation(s)
- Krithicaa Narayanaa Y
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Amrutha Arjunan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Devi Maigandan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Arun Dharmarajan
- Sri Ramachandra Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102 Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; School of Human Sciences, The University of Western Australia, Nedlands, Western Australia, Australia.
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| |
Collapse
|
199
|
Udofa E, Sankholkar D, Mitragotri S, Zhao Z. Antibody drug conjugates in the clinic. Bioeng Transl Med 2024; 9:e10677. [PMID: 39545074 PMCID: PMC11558205 DOI: 10.1002/btm2.10677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 11/17/2024] Open
Abstract
Antibody-drug conjugates (ADCs), chemotherapeutic agents conjugated to an antibody to enhance their targeted delivery to tumors, represent a significant advancement in cancer therapy. ADCs combine the precise targeting capabilities of antibodies and the potent cell-killing effects of chemotherapy, allowing for enhanced cytotoxicity to tumors while minimizing damage to healthy tissues. Here, we provide an overview of the current clinical landscape of ADCs, highlighting 11 U.S. Food and Drug Administration (FDA)-approved products and discussing over 500 active clinical trials investigating newer ADCs. We also discuss some key challenges associated with the clinical translation of ADCs and highlight emerging strategies to overcome these hurdles. Our discussions will provide useful guidelines for the future development of safer and more effective ADCs for a broader range of indications.
Collapse
Affiliation(s)
- Edidiong Udofa
- Department of Pharmaceutical SciencesUniversity of Illinois ChicagoChicagoIllinoisUSA
| | | | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesUniversity of Illinois ChicagoChicagoIllinoisUSA
- University of Illinois Cancer CenterChicagoIllinoisUSA
| |
Collapse
|
200
|
Zinovieva M, Ryapolova A, Karabelsky A, Minskaia E. Oncolytic Vesicular Stomatitis Virus: Optimisation Strategies for Anti-Cancer Therapies. FRONT BIOSCI-LANDMRK 2024; 29:374. [PMID: 39614430 DOI: 10.31083/j.fbl2911374] [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: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024]
Abstract
Oncolytic viruses (OVs) represent a targeted anti-cancer therapy approach due to their ability not only to selectively infect and destroy malignant cells but also to induce an immune response. Vesicular stomatitis virus (VSV) offers a promising platform due to its low prevalence and pathogenicity in humans, lack of pre-existing immunity, easily manipulated genome, rapid growth to high titers in a broad range of cell lines, and inability to integrate into the host genome. However, despite its many advantages, many unresolved problems remain: problematic production based on the reverse genetics system, oncological selectivity, and the overall effectiveness of VSV monotherapy. This review will discuss various attempts at viral genome modifications aimed at improving the oncolytic properties of VSV. These strategies include inhibition of viral genes, modification of genes responsible for targeting cancer cells over healthy ones, insertion of foreign genes for boosting immune response, and changing the order of viral and inserted foreign genes. In addition, possible ways to improve VSV-based anti-tumor therapy and achieve higher efficiency will be considered by evaluating the effectiveness of various delivery methods as well as discussing treatment options by combining VSV with other groups of anticancer drugs.
Collapse
Affiliation(s)
- Margarita Zinovieva
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|