1
|
Cerra B, Gioiello A. Discovery and development of steroidal enzyme inhibitors as anti-cancer drugs: state-of-the-art and future perspectives. J Enzyme Inhib Med Chem 2025; 40:2483818. [PMID: 40172115 PMCID: PMC11967001 DOI: 10.1080/14756366.2025.2483818] [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: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025] Open
Abstract
Steroidal compounds have emerged as effective therapeutic agents in oncology. Beyond natural-occurring and synthetic steroids that act as cytotoxic anti-tumoral agents, steroidal derivatives can be designed to mime the endogenous substrates of key metabolic enzymes in steroidogenesis, thus reducing the circulating levels of relevant oestrogenic and androgenic hormones responsible for cancer survival and proliferation. Therefore, enzyme inhibition represents an intriguing endocrine approach for the treatment of hormone-dependent tumours, such as breast and prostate cancer, with well-known approved drugs and several pre-clinical and clinical candidates under investigation. This review summarises the key advancements over the past decade (2014-2024) in the development of steroidal enzyme inhibitors endowed with anticancer activity, illustrating their mechanisms of action, therapeutic potential, drug design approaches, and current clinical applications. Furthermore, we discuss challenges related to drug resistance, off-target effects, and future strategies to optimise their efficacy in oncology.
Collapse
Affiliation(s)
- Bruno Cerra
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Antimo Gioiello
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Perumalsamy H, Sukweenadhi J, Ranjan A, Dubey A, Mahadev M, Elsadek MF, Almutairi SM, Sohn D, Balusamy SR. Structural isomers of carene persuade apoptotic cell death by inhibiting cell cycle in breast cancer cells: An in silico and in vitro approach. Tissue Cell 2025; 93:102701. [PMID: 39765134 DOI: 10.1016/j.tice.2024.102701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025]
Abstract
For the first time, our study provides a comprehensive examination of the anti-cancer effects of structural isomers of carene in breast cancer cells, specifically focusing on cell cycle inhibition and the induction of apoptosis. We utilized the hydro-distillation method to extract Piper nigrum seed essential oil (PNS-EO) and identified its bioactive components through gas chromatography-mass spectrometry (GC-MS) analysis. A total of 46 bioactive compounds were isolated via hydro-distillation, identified through GC-MS analysis, and validated by co-injection using GC analysis. The major constituent, 3-carene displayed the most substantial anti-proliferative effect on the breast cancer cell line MCF-7, with an IC50 value of 11.19 µg/mL. Further, docking studies were conducted to evaluate the putative role of 3-carene in inhibiting the cell cycle proteins (CDKN2A, CCND1, CDK4), as well as proteins in the apoptosis pathway (BCL-XL, BAX, BAK, Caspase 3). Additionally, we employed fluorescence-activated cell sorting (FACS) and clonogenic assays to evaluate cell cycle inhibition and time-dependent initiation of apoptosis. Moreover, fluorescence techniques including Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), Hoechst staining, and Propidium iodide (PI) staining were performed to assess cell death and apoptosis. Furthermore, molecular techniques such as quantitative real-time PCR (qPCR) and western blotting were utilized to investigate the mechanism of cell death was elucidated through the inhibition of Bcl-2, MMP2, MMP9, and Akt expression, alongside the activation of Bax, cytochrome C, and Caspases 3 and 9. Our findings indicate that 3-carene, isolated through hydro-distillation, effectively hinders the cell cycle and promotes apoptosis in MCF-7 cells. Consequently, it shows promise for incorporation into combinational anti-cancer therapies, warranting further research.
Collapse
Affiliation(s)
- Haribalan Perumalsamy
- Center for Creative Convergence Education, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea.
| | - Johan Sukweenadhi
- Center of Excellence for Food Products and Health Supplements for Degenerative Conditions, University of Surabaya, Surabaya 60293, Indonesia
| | - Anuj Ranjan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Sector 125, Noida, Uttar Pradesh, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru-575018, Karnataka, India
| | - Manohar Mahadev
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru-575018, Karnataka, India
| | - Mohamed Farouk Elsadek
- Department of Biochemistry, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Saeedah Musaed Almutairi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh 11451, Saudi Arabia
| | - Daewon Sohn
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea; Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-gu, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Ryszkiewicz P, Malinowska B, Schlicker E. Polypharmacology: new drugs in 2023-2024. Pharmacol Rep 2025:10.1007/s43440-025-00715-8. [PMID: 40095348 DOI: 10.1007/s43440-025-00715-8] [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: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Polypharmacology is an emerging approach to drug design and development that involves the use of multi-target-directed ligands (MTDLs), agents capable of interacting with multiple biological targets simultaneously. The effective treatment of chronic and multifactorial conditions, driven by the dysregulation of multiple interconnected pathways, such as cancer, autoimmune and metabolic disorders, cardiovascular and neurodegenerative diseases, is one of the most substantial challenges in contemporary pharmacology. 'Traditional' single-target-based treatment frequently shows limited effectiveness, as resistance to therapy develops or relapses occur. The rational use of MTDLs seems therefore a promising way to address the complexity of biological systems, feedback mechanisms, crosstalk, and molecular pathways. Many MTDLs have been successfully marketed to date. Moreover, plenty of them offer an additional benefit in comparison to 'traditional' treatment approaches. To assess whether the polypharmacological trend remains prevalent, we thoroughly analysed drugs approved in the years of 2023-2024 in Germany. Among 73 newly introduced substances, 18 are in line with the polypharmacology concept, including 10 antitumor agents, 5 drugs indicated for autoimmune disorders, 1 indicated for hand eczema, 1 antidiabetic (and anti-obesity) drug, and 1 modified corticosteroid.
Collapse
Affiliation(s)
- Piotr Ryszkiewicz
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Białystok, 15-222, Poland.
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Białystok, 15-222, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
4
|
Molefi T, Mabonga L, Hull R, Mwazha A, Sebitloane M, Dlamini Z. The Histomorphology to Molecular Transition: Exploring the Genomic Landscape of Poorly Differentiated Epithelial Endometrial Cancers. Cells 2025; 14:382. [PMID: 40072110 PMCID: PMC11898822 DOI: 10.3390/cells14050382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
The peremptory need to circumvent challenges associated with poorly differentiated epithelial endometrial cancers (PDEECs), also known as Type II endometrial cancers (ECs), has prompted therapeutic interrogation of the prototypically intractable and most prevalent gynecological malignancy. PDEECs account for most endometrial cancer-related mortalities due to their aggressive nature, late-stage detection, and poor response to standard therapies. PDEECs are characterized by heterogeneous histopathological features and distinct molecular profiles, and they pose significant clinical challenges due to their propensity for rapid progression. Regardless of the complexities around PDEECs, they are still being administered inefficiently in the same manner as clinically indolent and readily curable type-I ECs. Currently, there are no targeted therapies for the treatment of PDEECs. The realization of the need for new treatment options has transformed our understanding of PDEECs by enabling more precise classification based on genomic profiling. The transition from a histopathological to a molecular classification has provided critical insights into the underlying genetic and epigenetic alterations in these malignancies. This review explores the genomic landscape of PDEECs, with a focus on identifying key molecular subtypes and associated genetic mutations that are prevalent in aggressive variants. Here, we discuss how molecular classification correlates with clinical outcomes and can refine diagnostic accuracy, predict patient prognosis, and inform therapeutic strategies. Deciphering the molecular underpinnings of PDEECs has led to advances in precision oncology and protracted therapeutic remissions for patients with these untamable malignancies.
Collapse
Affiliation(s)
- Thulo Molefi
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
- Department of Medical Oncology, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Lloyd Mabonga
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| | - Absalom Mwazha
- Department of Anatomical Pathology, National Health Laboratory Services, Durban 4058, South Africa
| | - Motshedisi Sebitloane
- Discipline of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban 4002, South Africa;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP) Pan African Research Institute (PACRI), University of Pretoria, Hartfield, Pretoria 0028, South Africa
| |
Collapse
|
5
|
Pradhan B, Ki JS. Seaweed-derived laminarin and alginate as potential chemotherapeutical agents: An updated comprehensive review considering cancer treatment. Int J Biol Macromol 2025; 293:136593. [PMID: 39426775 DOI: 10.1016/j.ijbiomac.2024.136593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Seaweed-derived bioactive substances such as polysaccharides have proven to be effective chemotherapeutic and chemopreventive agents. Laminarin and alginate antioxidant properties aid in the prevention of cancer through dynamic modulation of critical intracellular signaling pathways via apoptosis which produce low cytotoxicity and potential chemotherapeutic effects. Understanding the effects of laminarin and alginate on human cancer cells and their molecular roles in cell death pathways can help to develop a novel chemoprevention strategy. This review emphasizes the importance of apoptosis-modulating laminarin and alginate in a range of malignancies as well as their extraction, molecular structure, and weight. In addition, future nano-formulation enhancements for greater clinical efficacy are discussed. Laminarin and alginate are perfect ingredients because of their distinct physicochemical and biological characteristics and their use-based delivery systems in cancer. The effectiveness of laminarin and alginate against cancer and more preclinical and clinical trials will open up as new chemotherapeutic natural drugs which lead to established as potential cancer drugs.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea; Department of Botany, Model Degree College, Rayagada 765017, Odisha, India
| | - Jang-Seu Ki
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea.
| |
Collapse
|
6
|
Yakkala PA, Kamal A. Dual-targeting inhibitors involving tubulin for the treatment of cancer. Bioorg Chem 2025; 156:108116. [PMID: 39823818 DOI: 10.1016/j.bioorg.2024.108116] [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: 10/05/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/20/2025]
Abstract
Combination therapies play a pivotal role in cancer treatment due to the intricate nature of the disease. Tubulin, a protein crucial for cellular functions, is a prime target in tumor therapy as it regulates microtubule dynamics. Combining tubulin inhibitors with other different inhibitors as dual targeting inhibitors has shown synergistic anti-tumor effects, amplifying therapeutic outcomes. Despite clinical approval of several tubulin inhibitors, their efficacy is hampered by drug resistance and toxic side effects. Dual targeting inhibitors of tubulin and other cancer-related pathways have emerged as vital components in cancer therapy, with promising prospects in both market availability and ongoing clinical trials. The rational design of hybrid inhibitors targeting both pathways presents an innovative approach to combatting cancer. However, despite the potent anti-tumor activity exhibited by several compounds, research on their anti-angiogenic potential remains limited. This review emphasizes the significance of tubulin based dual-target inhibitors, elucidating their mechanisms of action. Recent advances in exploring therapeutic efficacy, toxicity profiles, and challenges such as MDR are discussed. By presenting the research progress of tubulin based dual-target inhibitors as potential anticancer agents, this study delivers valuable insights for the development of more efficient drugs for cancer therapy.
Collapse
Affiliation(s)
- Prasanna Anjaneyulu Yakkala
- Department of Human Nutrition and Analytical Chemistry, Human Nutrition Program, The Ohio State University, Columbus, OH 43212, United States of America; Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ahmed Kamal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India; Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Dist. Medchal, 500078 TS, India.
| |
Collapse
|
7
|
Zhang J, Ren X, Song Y, Yu B. JBI-802: the first orally available LSD1/HDAC6 dual inhibitor to enter clinical trials. Expert Opin Ther Pat 2025:1-9. [PMID: 39960216 DOI: 10.1080/13543776.2025.2468792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
INTRODUCTION Lysine-specific demethylase 1 (LSD1) and histone deacetylase 6 (HDAC6) are key epigenetic regulators involved in histone demethylation and deacetylation processes that impact chromatin structure and gene expression. JBI-802 marks a major advancement as the first novel, orally available LSD1/HDAC6 dual inhibitor currently in clinical trials. AREAS COVERED This review provides a comprehensive overview of the discovery and development of JBI-802, detailing its structure-activity relationship (SARs), chemical synthesis, biological activity, and clinical progress. Other dual LSD1/HDAC6 inhibitors and the challenges are briefly discussed, underscoring the therapeutic potential of dual inhibition in disease treatment. The literature search is performed using SciFinder, Google patent, ClinicalTrials databases, and PubMed. EXPERT OPINION The dual LSD1/HDAC6 inhibitor JBI-802 demonstrates robust anti-proliferative activity, significant antitumor effects in multiple hematologic malignancies, and superior efficacy in combination with checkpoint inhibitors in the CT-26 syngeneic mouse model. JBI-802 is currently undergoing phase I/II clinical trials in patients with advanced solid tumors, myeloproliferative neoplasms (MPN), and myelodysplastic/myeloproliferative neoplasms (MDS/MPN) with thrombocytosis. However, the potential on-target toxicity, off-target interactions and selectivity concerns deservee more attention.
Collapse
Affiliation(s)
- Jingya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiangli Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, Fujian Medical University, Fuzhou, China
| | - Bin Yu
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Nafie MS, Fahmy SA, Kahwash SH, Diab MK, Dawood KM, Abbas AA. Recent advances on anticancer activity of benzodiazine heterocycles through kinase inhibition. RSC Adv 2025; 15:5597-5638. [PMID: 39974315 PMCID: PMC11836603 DOI: 10.1039/d4ra08134j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
The benzodiazines (phthalazine, quinazoline, quinoxaline, and cinnoline) have emerged as attractive scaffolds for creating novel anticancer drugs. These nitrogen-containing heterocycles are intriguing because they have a variety of configurations and can change chemically, allowing us to tailor their pharmacokinetic and pharmacodynamic features. Numerous studies have found that derivatives of these compounds have potent anticancer properties via inhibiting topoisomerases, protein kinases, and receptor tyrosine kinases. These compounds impair critical processes that control cancer proliferation and survival. Most benzodiazine derivatives have achieved clinical success, demonstrating the heterocycles' therapeutic potential. The use of phthalazine, cinnoline, and quinazoline derivatives should open new avenues in developing better and more targeted cancer treatments. In this overview, we summarize recent advances in synthesizing these compounds and illustrate how they serve as promising chemotherapeutic agents. Therefore, current research organizes the latest information to provide a clearer picture of design strategies that boost efficacy and selectivity, allowing the identification of potential anticancer drug candidates down the line. This research study also highlights the need to establish heterocyclic derivatives as a promising source of new molecules for cancer treatment with improved efficacy and decreased effects.
Collapse
Affiliation(s)
- Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah Sharjah 27272 United Arab Emirates
- Department of Chemistry, Faculty of Science, Suez Canal University Ismailia 41522 Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Shaima H Kahwash
- Department of Chemistry, Faculty of Science, Suez Canal University Ismailia 41522 Egypt
| | - Mohamed K Diab
- Pest Physiology Department, Plant Protection Research Institute, Agricultural Research Center Giza 12311 Egypt
| | - Kamal M Dawood
- Chemistry Department, Faculty of Science, Cairo University Giza 12613 Egypt
| | - Ashraf A Abbas
- Chemistry Department, Faculty of Science, Cairo University Giza 12613 Egypt
| |
Collapse
|
9
|
Effiong ME, Bella-Omunagbe M, Afolabi IS, Chinedu SN. Molecular Docking Appraisal of Pleurotus ostreatus Phytochemicals as Potential Inhibitors of PI3K/Akt Pathway for Breast Cancer Treatment. Bioinform Biol Insights 2025; 19:11779322251316864. [PMID: 39906062 PMCID: PMC11792010 DOI: 10.1177/11779322251316864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
Introduction Breast cancer (BC) is a heterogeneous disease involving a network of numerous extracellular signal transduction pathways. The phosphoinositide 3-kinase (PI3K)/serine/threonine kinase (Akt)/mechanistic target of rapamycin (mTOR) pathway is crucial for understanding the BC development. Phosphoinositide 3-kinase, phosphatase and tensin homolog (PTEN), mTOR, Akt, 3-phosphoinositide-dependent kinase 1 (PDK1), FoxO1, glycogen synthase kinase 3 (GSK-3), mouse double minute 2 (MDM2), H-Ras, and proapoptotic B-cell lymphoma 2 (BCL-2) family protein (BAD) proteins are key drivers of this pathway and potential therapeutic targets. Pleurotus ostreatus is an edible mushroom that is rich in flavonoids and phenols that can serve as potential inhibitors of proteins in the PI3K/Akt/mTOR pathway. Aim This study evaluated the anticancer properties of P ostreatus through a structure-based virtual screening of 22 biologically active compounds present in the mushroom. Method Model optimization was carried out on PI3K, PTEN, mTOR, Akt, PDK1, FoxO1, GSK-3, MDM2, H-Ras, and BAD proteins in the PI3K/Akt/mTOR pathway and molecular docking of compounds/control inhibitors in the binding pocket were simulated AutoDock Vina in PyRx. The drug likeness, pharmacokinetic, and pharmacodynamic features of prospective docking leads were all anticipated. Result Several potent inhibitors of the selected key driver proteins in PI3K/Akt/mTOR pathway were identified from P ostreatus. Ellagic acid with binding affinities of -8.0, -8.0, -8.1, -8.2, -6.2, and -7.1 kcal/mol on PI3K, Akt, PDK1, GSK-3, MDM2, and BAD, respectively, had better binding affinity compared with their reference drugs. Likewise, apigenin (-7.8 kcal/mol), chrysin (-7.8 kcal/mol), quercetin (-6.4 kcal/mol), and chlorogenic acid (-6.2 kcal/mol) had better binding affinities to PTEN, mTOR, FoxO1, and H-Ras proteins, respectively. Conclusion Ellagic acid, apigenin, luteolin, quercetin, chlorogenic acid, chrysin, and naringenin phytochemicals are seen as the better lead molecules due to their ability to strongly bind to the proteins under study in this pathway. Analogs of these compounds can also be designed as potential drugs.
Collapse
Affiliation(s)
- Magdalene Eno Effiong
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Mercy Bella-Omunagbe
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Israel Sunmola Afolabi
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, Ota, Nigeria
| | - Shalom Nwodo Chinedu
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, Ota, Nigeria
| |
Collapse
|
10
|
Gui H, Nie Y, Yuan H, Wang M, Li L, Zhu L, Chen S, Jing Q, Wan Q, Lv H, Nie Y, Zhang X. Ansofaxine suppressed NSCLC progression by increasing sensitization to combination immunotherapy. Int Immunopharmacol 2025; 146:113918. [PMID: 39718058 DOI: 10.1016/j.intimp.2024.113918] [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: 09/21/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Depression negatively impacts the prognosis of various cancers, including lung cancer, by influencing antitumor immune responses and impairing immune cell function. Antidepressants may modulate the tumor immune microenvironment, enhancing immunotherapy efficacy. However, the specific mechanisms remain unclear. This study investigates the effects of the antidepressant Ansofaxine on immune therapy in non-small cell lung cancer (NSCLC) mice with comorbid depression. METHODS Chronic unpredictable mild stress (CUMS) and Lewis lung cancer cells (LLC) model was established in mice. Ansofaxine and a combination of triple immunotherapy (anti-PD-1, anti-TNFR2, and anti-PTP1B) were treated in mice to monitor tumor growth and survival rates. Flow cytometry and immunohistochemistry were employed to analyze the dynamics of the immune system, while ELISA kits were used to quantify neurotransmitter levels. RESULTS Depression accelerated NSCLC progression, evidenced by increased tumor volume, spleen size, and reduced survival rates. Flow cytometry analysis demonstrated a reduction in the population of immune effector cells, with an increase in the proportion of immunosuppressive cells. Ansofaxine inhibited LLC cell proliferation and migration, enhancing apoptosis more effectively than venlafaxine and fluoxetine. Combined with triple immunotherapy, Ansofaxine improved survival rates and enhanced immune responses, increasing CD8+ T cell proportions and decreasing Tregs. Ansofaxine also restored serum serotonin and norepinephrine levels in depressed mice, reduced corticosterone, and decreased PD-L1 and TNFR2 expression in tumor tissues. CONCLUSION The findings suggest that Ansofaxine may represent a promising therapeutic approach for NSCLC patients with comorbid depression, potentially enhancing both mental well-being and cancer-related outcomes.
Collapse
Affiliation(s)
- Huan Gui
- School of Medicine, Guizhou University, Guiyang 550025, China; Department of Hyperbaric Oxygen, People's Hospital of Qianxinan Prefecture, Xingyi 562400, China
| | - Yujie Nie
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Haohua Yuan
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Mengjiao Wang
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Linzhao Li
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Lan Zhu
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Shuanghui Chen
- School of Medicine, Guizhou University, Guiyang 550025, China
| | - Qianyu Jing
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Quan Wan
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Hang Lv
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yingjie Nie
- School of Medicine, Guizhou University, Guiyang 550025, China; Shenzhen Hospital, The University of Hongkong, Shenzhen 518053, China.
| | - Xiangyan Zhang
- School of Medicine, Guizhou University, Guiyang 550025, China; NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| |
Collapse
|
11
|
Baroni C, Bozdag M, Renzi G, De Luca V, Capasso C, Bazzicalupi C, Selleri S, Ferraroni M, Carta F, Supuran CT. X-ray crystallographic and kinetic studies of biguanide containing aryl sulfonamides as carbonic anhydrase inhibitors. RSC Med Chem 2025:d4md01018c. [PMID: 39935522 PMCID: PMC11809658 DOI: 10.1039/d4md01018c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Here, we report a small series of dual-targeting compounds that combine the prototypical carbonic anhydrase (CA) zinc-binding sulfonamide moiety with the biguanide group of metformin, an emerging anticancer drug. The compounds reported similar in vitro inhibition profiles on a panel of physiologically relevant human (h)CAs, with marked selectivity for the cancer related IX and XII isoforms. The binding modes of representative inhibitors 5b and 5c within the active site of the hCA isoforms II and XII-mimic were assessed by X-ray crystallography, thus allowing us to clarify molecular features that may be useful for the design of more specific and potent inhibitors. For instance, we identified a mutation in the hCA XII-mimic which was found responsible for the selectivity of the ligands toward the tumor associated isoform. Interestingly, in the hCA II/5c complex, a second inhibitor molecule was bound to the catalytic cleft, probably affecting the inhibition properties of the canonical zinc-bound inhibitor.
Collapse
Affiliation(s)
- Chiara Baroni
- Department of Chemistry "Ugo Schiff", University of Florence Via della Lastruccia 3 50019 Sesto Fiorentino FI Italy
| | - Murat Bozdag
- NEUROFARBA Department, University of Florence Via Ugo Schiff 6 50019 Sesto Fiorentino FI Italy
| | - Gioele Renzi
- NEUROFARBA Department, University of Florence Via Ugo Schiff 6 50019 Sesto Fiorentino FI Italy
| | - Viviana De Luca
- Department of Biology, Institute of Bioscience and Bioresources (IBBR)-CNR Via P. Castellino 111 80131 Napoli NA Italy
| | - Clemente Capasso
- Department of Biology, Institute of Bioscience and Bioresources (IBBR)-CNR Via P. Castellino 111 80131 Napoli NA Italy
| | - Carla Bazzicalupi
- Department of Chemistry "Ugo Schiff", University of Florence Via della Lastruccia 3 50019 Sesto Fiorentino FI Italy
| | - Silvia Selleri
- NEUROFARBA Department, University of Florence Via Ugo Schiff 6 50019 Sesto Fiorentino FI Italy
| | - Marta Ferraroni
- Department of Chemistry "Ugo Schiff", University of Florence Via della Lastruccia 3 50019 Sesto Fiorentino FI Italy
| | - Fabrizio Carta
- NEUROFARBA Department, University of Florence Via Ugo Schiff 6 50019 Sesto Fiorentino FI Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, University of Florence Via Ugo Schiff 6 50019 Sesto Fiorentino FI Italy
| |
Collapse
|
12
|
Badawi WA, Okda TM, Abd El Wahab SM, Ezz-ElDien ES, AboulWafa OM. Developing new anticancer agents: Design, synthesis, biological evaluation and in silico study of several functionalized pyrimidine-5-carbonitriles as small molecules modulators targeting breast cancer. Bioorg Chem 2024; 153:107953. [PMID: 39556931 DOI: 10.1016/j.bioorg.2024.107953] [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: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024]
Abstract
Committed to our growing effort addressed toward the development of potent anti-breast cancer candidates, new 4-hydrazinylpyrimidine-5-carbonitriles featuring a morpholinyl or piperidinyl moiety at the position-2 and derivatized with various functionalities at the hydrazinyl group were designed through structure optimization, and their antiproliferative potency against two human breast cancer (BC) cell lines, relative to the reference drug 5-FU, was evaluated. Compounds showing remarkable cytotoxic activity versus the hormone dependent MCF-7 cell line (IC50 = 1.62 ± 0.06 µM- 9.88 ± 0.38 µM) and the non-hormone dependent MDA-MB-231 cell line (IC50 = 3.26 ± 0.14 µM-12.93 ± 0.55 µM) were further tested by multiple assays for clarification of their potential activity. Promising derivatives revealing low damage to healthy cells were subject to enzymatic inhibitory assessment against ARO and EGFR and their activities compared to letrozole and erlotinib respectively. Compounds 3c, 6a as well as compounds 4c, 4d proved to be good inhibitors of the ARO and EGFR enzymes respectively. Active compounds were also evaluated for their underlying mode of action by further investigation for CDK, Hsp90, PI3K inhibition and compared to normal MCF-10A cells and assessed for their enhancement of the caspase 9 levels. Additionally, cell cycle analysis and apoptotic induction were performed. They demonstrated remarkable activities in the previous assays and emanated as leads as anti-breast cancer candidates. Eventually, molecular docking analysis revealed that hit compounds 3c, 4c, 4d, and 6a could bind favorably to the proposed in silico models of various protein-ligand interactions. Therefore, our promising top candidates, by demonstrating appreciable anti-breast cancer activities, present valuable prospects for optimization, potency enhancement and future application.
Collapse
Affiliation(s)
- Waleed A Badawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Damanhour, 22511, Egypt.
| | - Tarek M Okda
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Shrouk M Abd El Wahab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Damanhour, 22511, Egypt
| | - Eman S Ezz-ElDien
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Damanhour, 22511, Egypt
| | - Omaima M AboulWafa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21215, Egypt
| |
Collapse
|
13
|
Ali BS, Mohammed AF, Kariuki BM, El-Awady R, H M Abdu-Allah H. Tetrahydrocarbazoles incorporating 5-arylidene-4-thiazolinones as potential antileukemia and antilymphoma targeting tyrosine kinase and tubulin polymerase enzymes: Design, synthesis, structural, biological and molecular docking studies. Bioorg Chem 2024; 153:107817. [PMID: 39278066 DOI: 10.1016/j.bioorg.2024.107817] [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: 07/11/2024] [Revised: 08/23/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
Finding effective and selective anticancer agents is a top medical priority due to high clinical treatment demand. However, current anticancer agents have serious side effects and resistance development remains a big concern. This creates an urgent need for new multitarget drugs that could solve these problems. Tetrahydrocarbazoles and 5-arylidene-4-thiazolinones have always attracted researchers for their multifaced anticancer activities and the possibility to be easily derivatized. Thereby, herein we report the combination of the two scaffolds to provide compounds 9a-j and 10a-j that were fully characterized and their tautomeric form was confirmed by crystal structure. 9a-j and 10a-j wereassessedfor invitro antiproliferative activityusing SRB assay against a panel of seven human cancer cell lines with doxorubicin as the standard. The results revealed that the cell lines derived from leukemia (Jurkat) and lymphoma (U937) are the most sensitive. Compounds 9d, 10e, 10g, and 10f revealed the highest potency (IC50 = 3.11-11.89 μM) with much lower effects on normal lymphocytes cell line (IC50 > 50 µM). The results show that modifications at 6th position of the THC and the nature of the substituent at the arylidene moiety affect the activity. To exploit the mode of action, 9d, 10e, 10f, and 10g were evaluated as VEGFR-2 and EGFR inhibitors. 10e is the most potent (IC50 0.26 and 0.14 μM) against both enzymes. It also induced G0-G1-phase cell cycle arrest and apoptosis. While 10g exhibited higher potency (IC50 9.95 μM) than vincristine (IC50 15.63 μM) against tubulin. A molecular docking study was carried out to understand the interactions between 10e, 10g and their targets. This study reveals 10e and 10g as possible candidates for developing multitarget anticancer agents against leukemia and lymphoma.
Collapse
Affiliation(s)
- Basma S Ali
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Anber F Mohammed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | | | - Raafat El-Awady
- Research Institute for Medical and Health Sciences and College of Pharmacy, University of sharjah, Sharjah 27272, United Arab Emirates
| | - Hajjaj H M Abdu-Allah
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| |
Collapse
|
14
|
Alharthi F, Althagafi HA, Jafri I, Oyouni AAA, Althaqafi MM, Al-Hazmi NE, Al Hijab LYA, Naguib DM. Enhancing the Stability and Anticancer Activity of Escherichia coli Asparaginase Through Nanoparticle Immobilization: A Biotechnological Perspective on Nano Chitosan. Polymers (Basel) 2024; 16:3260. [PMID: 39684005 DOI: 10.3390/polym16233260] [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: 09/29/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
There is a shortage in the experimental research directly comparing the effectiveness of different nanoparticles in boosting asparaginase (ASNase) activity. This study assessed the impact of various nanoparticles on enhancing ASNase activity, stability, and anticancer effects through immobilization. Escherichia coli ASNase was immobilized on different nanoparticles, and its efficiency was measured. The research included analyzing the enzyme's secondary structure, stability, activity at different temperatures, kinetic parameters, shelf life, and activity in blood serum. The anticancer efficacy was determined by measuring the IC50. The study also investigated the anticancer mechanisms by examining the enzyme's toxicity on cancer cells, focusing on apoptosis indicators like nuclear intensity, membrane permeability, mitochondrial membrane permeability, and cytochrome c release. Among the tested nanoparticles, nano chitosan yielded the best improvements. ASNase immobilized on nano chitosan reached 90% immobilization efficiency fastest among the studied nanoparticles, achieving this within 72 h, whereas other nanoparticles took 120 h. Immobilization modified ASNase's secondary structure by increasing alpha helices and reducing random coils, with nanochitosan and magnetic iron oxide showing the most pronounced effects. Immobilized ASNase exhibited enhanced activity, stability across temperature (widest with nanochitosan, 25-65 °C), and a broader optimal pH range compared to the free enzyme, with a Km of 1.227 mM and a Vmax of 454.54 U/mg protein. Notably, the nano-chitosan-immobilized ASNase retained over 85% of its activity after 9 months of storage and maintained high activity in blood serum. This improved stability and activity translated into the highest anticancer activity (Lowest IC50) and was more effective than doxorubicin in disrupting cancer cell structures.
Collapse
Affiliation(s)
- Fahad Alharthi
- Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hussam A Althagafi
- Department of Biology, Faculty of Science, Al-Baha University, Al-Baha 65525, Saudi Arabia
| | - Ibrahim Jafri
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Atif Abdulwahab A Oyouni
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
- Biodiversity Genomics Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammed M Althaqafi
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Nawal E Al-Hazmi
- Department of Chemistry, Division of Biology (Microbiology), University College of Qunfudah, Umm Al-Qura University, Qunfudah 21961, Saudi Arabia
| | | | - Deyala M Naguib
- Department of Biology, Faculty of Science, Al-Baha University, Al-Baha 65525, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
15
|
Zhu B, Xiang K, Li T, Li X, Shi F. The signature of extracellular vesicles in hypoxic breast cancer and their therapeutic engineering. Cell Commun Signal 2024; 22:512. [PMID: 39434182 PMCID: PMC11492701 DOI: 10.1186/s12964-024-01870-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024] Open
Abstract
Breast cancer (BC) currently ranks second in the global cancer incidence rate. Hypoxia is a common phenomenon in BC. Under hypoxic conditions, cells in the tumor microenvironment (TME) secrete numerous extracellular vesicles (EVs) to achieve intercellular communication and alter the metabolism of primary and metastatic tumors that shape the TME. In addition, emerging studies have indicated that hypoxia can promote resistance to tumor treatment. Engineered EVs are expected to become carriers for cancer treatment due to their high biocompatibility, low immunogenicity, high drug delivery efficiency, and ease of modification. In this review, we summarize the mechanisms of EVs in the primary TME and distant metastasis of BC under hypoxic conditions. Additionally, we highlight the potential applications of engineered EVs in mitigating the malignant phenotypes of BC cells under hypoxia.
Collapse
Affiliation(s)
- Baiheng Zhu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kehao Xiang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Tanghua Li
- The First Clinical Medical School, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Fujun Shi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
16
|
Ardini M, Aboagye SY, Petukhova VZ, Kastrati I, Ippoliti R, Thatcher GRJ, Petukhov PA, Williams DL, Angelucci F. The "Doorstop Pocket" In Thioredoxin Reductases─An Unexpected Druggable Regulator of the Catalytic Machinery. J Med Chem 2024; 67:15947-15967. [PMID: 39250602 DOI: 10.1021/acs.jmedchem.4c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Pyridine nucleotide-disulfide oxidoreductases are underexplored as drug targets, and thioredoxin reductases (TrxRs) stand out as compelling pharmacological targets. Selective TrxR inhibition is challenging primarily due to the reliance on covalent inhibition strategies. Recent studies identified a regulatory and druggable pocket in Schistosoma mansoni thioredoxin glutathione reductase (TGR), a TrxR-like enzyme, and an established drug target for schistosomiasis. This site is termed the "doorstop pocket" because compounds that bind there impede the movement of an aromatic side-chain necessary for the entry and exit of NADPH and NADP+ during enzymatic turnover. This discovery spearheaded the development of new TGR inhibitors with efficacies surpassing those of current schistosomiasis treatment. Targeting the "doorstop pocket" is a promising strategy, as the pocket is present in all members of the pyridine nucleotide-disulfide oxidoreductase family, opening new avenues for exploring therapeutic approaches in diseases where the importance of these enzymes is established, including cancer and inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Sammy Y Aboagye
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Valentina Z Petukhova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Irida Kastrati
- Department of Cancer Biology, Loyola University Chicago, 60153 Maywood, Illinois 60153, United States
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Gregory R J Thatcher
- Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Pavel A Petukhov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - David L Williams
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
17
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [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: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
18
|
Rogovskii V. Tumor-produced immune regulatory factors as a therapeutic target in cancer treatment. Front Immunol 2024; 15:1416458. [PMID: 39206193 PMCID: PMC11349530 DOI: 10.3389/fimmu.2024.1416458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Vladimir Rogovskii
- Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
19
|
Hao Y, Li B, Huang D, Wu S, Wang T, Fu L, Liu X. Developing a Semi-Supervised Approach Using a PU-Learning-Based Data Augmentation Strategy for Multitarget Drug Discovery. Int J Mol Sci 2024; 25:8239. [PMID: 39125808 PMCID: PMC11312053 DOI: 10.3390/ijms25158239] [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: 06/21/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Multifactorial diseases demand therapeutics that can modulate multiple targets for enhanced safety and efficacy, yet the clinical approval of multitarget drugs remains rare. The integration of machine learning (ML) and deep learning (DL) in drug discovery has revolutionized virtual screening. This study investigates the synergy between ML/DL methodologies, molecular representations, and data augmentation strategies. Notably, we found that SVM can match or even surpass the performance of state-of-the-art DL methods. However, conventional data augmentation often involves a trade-off between the true positive rate and false positive rate. To address this, we introduce Negative-Augmented PU-bagging (NAPU-bagging) SVM, a novel semi-supervised learning framework. By leveraging ensemble SVM classifiers trained on resampled bags containing positive, negative, and unlabeled data, our approach is capable of managing false positive rates while maintaining high recall rates. We applied this method to the identification of multitarget-directed ligands (MTDLs), where high recall rates are critical for compiling a list of interaction candidate compounds. Case studies demonstrate that NAPU-bagging SVM can identify structurally novel MTDL hits for ALK-EGFR with favorable docking scores and binding modes, as well as pan-agonists for dopamine receptors. The NAPU-bagging SVM methodology should serve as a promising avenue to virtual screening, especially for the discovery of MTDLs.
Collapse
Affiliation(s)
- Yang Hao
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Bo Li
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Daiyun Huang
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- School of Life Sciences, Fudan University, Shanghai 200092, China
| | - Sijin Wu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| | - Tianjun Wang
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZX, UK
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| | - Xin Liu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China; (Y.H.); (B.L.); (S.W.); (T.W.); (L.F.)
| |
Collapse
|
20
|
Huang P, Wen F, Li Q. Current concepts of the crosstalk between lncRNA and E2F1: shedding light on the cancer therapy. Front Pharmacol 2024; 15:1432490. [PMID: 39119602 PMCID: PMC11306149 DOI: 10.3389/fphar.2024.1432490] [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: 05/14/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) constitute a distinctive subset of RNA molecules with limited protein-coding potential, which exert crucial impacts on various biological activities. In the context of cancer, dysregulated lncRNAs function as essential regulators that affect tumor initiation and malignant progression. These lncRNAs serve as competitive endogenous RNAs (ceRNAs) through sponging microRNAs and regulating the expression of targeted genes. Moreover, they also directly bind to RNA-binding proteins, which can be integrated into a complex mechanistic network. E2F1, an extensively studied transcription factor, mediates multiple malignant behaviors by regulating cell cycle progression, tumor metastasis, and therapeutic response. Emerging evidence suggests that lncRNAs play a pivotal role in regulating the E2F1 pathway. This review aims to elucidate the intricate gene regulatory programs between lncRNAs and E2F1 in cancer progression. We elaborate on distinct mechanistic networks involved in cancer progression, emphasizing the potential of the lncRNAs/E2F1 axes as promising targets for cancer therapy. Additionally, we provide novel perspectives on current evidence, limitations, and future directions for targeting lncRNAs in human cancers. Fully deciphering the intricate network of lncRNA/E2F1-mediated regulatory mechanisms in cancer could facilitate the translation of current findings into clinical course, such efforts ultimately significantly improve the clinical prognosis of cancer patients.
Collapse
Affiliation(s)
- Peng Huang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Wen
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiu Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Marchesi E, Melloni E, Casciano F, Pozza E, Argazzi R, De Risi C, Preti L, Perrone D, Navacchia ML. Evaluation of Anticancer Activity of Nucleoside-Nitric Oxide Photo-Donor Hybrids. Molecules 2024; 29:3383. [PMID: 39064961 PMCID: PMC11279448 DOI: 10.3390/molecules29143383] [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: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Herein, we report the synthesis of a new hybrid compound based on a 2'-deoxyuridine nucleoside conjugated with a NO photo-donor moiety (dU-t-NO) via CuAAC click chemistry. Hybrid dU-t-NO, as well as two previously reported 2'-deoxyadenosine based hybrids (dAdo-S-NO and dAdo-t-NO), were evaluated for their cytotoxic and cytostatic activities in selected cancer cell lines. dAdo-S-NO and dAdo-t-NO hybrids displayed higher activity with respect to dU-t-NO. All hybrids showed effective release of NO in the micromolar range. The photochemical behavior of the newly reported hybrid, dU-t-NO, was studied in the RKO colon carcinoma cell line, whereas the dAdo-t-NO hybrid was tested in both colon carcinoma RKO and hepatocarcinoma Hep 3B2.1-7 cell lines to evaluate the potential effect of NO released upon irradiation on cell viability. A customized irradiation apparatus for in vitro experiments was also designed.
Collapse
Affiliation(s)
- Elena Marchesi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
| | - Elisabetta Melloni
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Elena Pozza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (F.C.); (E.P.)
| | - Roberto Argazzi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy
| | - Carmela De Risi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (E.M.); (R.A.); (C.D.R.)
| | - Lorenzo Preti
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Daniela Perrone
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Maria Luisa Navacchia
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy
| |
Collapse
|
22
|
López-López E, Medina-Franco JL. Toward structure-multiple activity relationships (SMARts) using computational approaches: A polypharmacological perspective. Drug Discov Today 2024; 29:104046. [PMID: 38810721 DOI: 10.1016/j.drudis.2024.104046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
In the current era of biological big data, which are rapidly populating the biological chemical space, in silico polypharmacology drug design approaches help to decode structure-multiple activity relationships (SMARts). Current computational methods can predict or categorize multiple properties simultaneously, which aids the generation, identification, curation, prioritization, optimization, and repurposing of molecules. Computational methods have generated opportunities and challenges in medicinal chemistry, pharmacology, food chemistry, toxicology, bioinformatics, and chemoinformatics. It is anticipated that computer-guided SMARts could contribute to the full automatization of drug design and drug repurposing campaigns, facilitating the prediction of new biological targets, side and off-target effects, and drug-drug interactions.
Collapse
Affiliation(s)
- Edgar López-López
- Department of Chemistry and Graduate Program in Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, Section 14-740, Mexico City 07000, Mexico; DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| | - José L Medina-Franco
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| |
Collapse
|