101
|
Chitluri KK, Emerson IA. The importance of protein domain mutations in cancer therapy. Heliyon 2024; 10:e27655. [PMID: 38509890 PMCID: PMC10950675 DOI: 10.1016/j.heliyon.2024.e27655] [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: 10/11/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
Cancer is a complex disease that is caused by multiple genetic factors. Researchers have been studying protein domain mutations to understand how they affect the progression and treatment of cancer. These mutations can significantly impact the development and spread of cancer by changing the protein structure, function, and signalling pathways. As a result, there is a growing interest in how these mutations can be used as prognostic indicators for cancer prognosis. Recent studies have shown that protein domain mutations can provide valuable information about the severity of the disease and the patient's response to treatment. They may also be used to predict the response and resistance to targeted therapy in cancer treatment. The clinical implications of protein domain mutations in cancer are significant, and they are regarded as essential biomarkers in oncology. However, additional techniques and approaches are required to characterize changes in protein domains and predict their functional effects. Machine learning and other computational tools offer promising solutions to this challenge, enabling the prediction of the impact of mutations on protein structure and function. Such predictions can aid in the clinical interpretation of genetic information. Furthermore, the development of genome editing tools like CRISPR/Cas9 has made it possible to validate the functional significance of mutants more efficiently and accurately. In conclusion, protein domain mutations hold great promise as prognostic and predictive biomarkers in cancer. Overall, considerable research is still needed to better define genetic and molecular heterogeneity and to resolve the challenges that remain, so that their full potential can be realized.
Collapse
Affiliation(s)
- Kiran Kumar Chitluri
- Bioinformatics Programming Lab, Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| | - Isaac Arnold Emerson
- Bioinformatics Programming Lab, Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN, 632014, India
| |
Collapse
|
102
|
Mazzoleni B, Ferrari G, Savioni F, Gravante F, Lopane D, Dacomi A, Coldani C, Tomaiuolo G, Cattani D, Anastasi G, Mancin S. Non-pharmacological strategies to alleviate dysgeusia in patients undergoing chemotherapy: A systematic review. Eur J Oncol Nurs 2024; 70:102569. [PMID: 38593535 DOI: 10.1016/j.ejon.2024.102569] [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: 02/10/2024] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE Dysgeusia is a common side effect in oncology patients, significantly impacting their quality of life. This systematic review aims to evaluate the effectiveness of non-pharmacological strategies in treating dysgeusia in patients undergoing chemotherapy or radiotherapy. METHODS Adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a comprehensive literature search across five databases: PubMed, Embase, Cochrane Library, CINAHL, and the British Nursing Database. We used the Joanna Briggs Institute Critical Appraisal Tools to assess the quality of the included studies. A harvest plot was used to synthesise evidence about the differential effects of population-level interventions. RESULTS Nine studies of non-pharmacological strategies to manage dysgeusia were included. These studies encompassed a variety of interventions, including oral applications and supplements, instrumental techniques, and educational programs. The review identified promising interventions such as cryotherapy and Miraculine supplementation, which showed potential in mitigating taste alterations. Instrumental techniques like photobiomodulation therapy and complementary and integrative medicine approaches, including acupuncture and herbs, were also found to be beneficial. Educational and self-management strategies emerged as effective interventions for empowering patients to manage dysgeusia. Despite the diversity of interventions and the limitations of the included studies, such as small sample sizes and geographical differences, these findings underscore the potential of non-pharmacological strategies in managing dysgeusia. CONCLUSION The results support the integration of these strategies into clinical practice, highlighting the importance of multidisciplinary approaches to improve patient care. Further research should prioritize rigorous studies to enhance evidence and explore long-term effects.
Collapse
Affiliation(s)
- Beatrice Mazzoleni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Giulia Ferrari
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Francesco Gravante
- Department of Anaesthesiology, Intensive Care Unit, Local Health Autority of Caserta, Caserta, Italy
| | - Diego Lopane
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alessandra Dacomi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Chiara Coldani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppina Tomaiuolo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Daniela Cattani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuliano Anastasi
- Department of Trauma, AOU G. Martino University Hospital, Messina, Italy
| | - Stefano Mancin
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
103
|
Moulana MS, Haiaty S, Bazmani A, Shabkhizan R, Moslehian MS, Sadeghsoltani F, Mostafazadeh M, Asadi MR, Talebi M, Jafari Z, Morovati MR, Farzaei MH, Rahbarghazi R. Tumoricidal properties of thymoquinone on human colorectal adenocarcinoma cells via the modulation of autophagy. BMC Complement Med Ther 2024; 24:132. [PMID: 38532470 DOI: 10.1186/s12906-024-04432-2] [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: 09/15/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Colorectal cancer (CRC) is deadly anaplastic changes in the gastrointestinal tract with high-rate mortality. In recent years, the application of phytocompounds has been extended along with different therapeutic protocols. Here, we monitored the effects of Thymoquinone (TQ) on autophagy via mitochondrial function after modulation of the Wnt/β-catenin signaling pathway.Human colorectal adenocarcinoma HT-29 cells were treated with TQ (60 µM) and 15 µM Wnt3a inhibitor (LGK974) for 48 h. The survival rate was evaluated using an MTT assay. The expression of Wnt-related factors (c-Myc, and Axin), angiogenesis (VE-Cadherin), and mitophagy-related factors (PINK1, OPTN) was assessed using real-time PCR assay. Protein levels of autophagy factors (Beclin-1, LC3, and P62) were monitored using western blotting. Using flow cytometry analysis, the intracellular accumulation of Rhodamine 123 was evaluated. The migration properties were analyzed using a scratch wound healing assay.Data indicated that TQ can reduce the viability of HT-29 cells compared to the control cells (p < 0.05). The expression of VE-Cadherin was inhibited while the expression of PINK1 was induced in treated cells (p < 0.05). Both LGK974 and TQ-treated cells exhibited activation of autophagy flux (Beclin-1↑, LC3II/I↑, and p62↓) compared to the control group (p < 0.05). TQ can increase intracellular accumulation of Rhodamine 123, indicating the inhibition of efflux mechanisms in cancer cells. Along with these changes, the migration of cells was also reduced (p < 0.05).TQ is a potential phytocompound to alter the dynamic growth of human colorectal HT-29 cells via the modulation of autophagy, and mitophagy-related mechanisms.
Collapse
Affiliation(s)
- Mohammad Saleh Moulana
- Department of Persian Medicine, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sanya Haiaty
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Bazmani
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Shabkhizan
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Sadat Moslehian
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Mostafazadeh
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Asadi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Jafari
- Department of Persian Medicine, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Morovati
- Department of Persian Medicine, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Traditional Pharmacy, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
104
|
Negut I, Bita B. Polymersomes as Innovative, Stimuli-Responsive Platforms for Cancer Therapy. Pharmaceutics 2024; 16:463. [PMID: 38675124 PMCID: PMC11053450 DOI: 10.3390/pharmaceutics16040463] [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: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This review addresses the urgent need for more targeted and less toxic cancer treatments by exploring the potential of multi-responsive polymersomes. These advanced nanocarriers are engineered to deliver drugs precisely to tumor sites by responding to specific stimuli such as pH, temperature, light, hypoxia, and redox conditions, thereby minimizing the side effects associated with traditional chemotherapy. We discuss the design, synthesis, and recent applications of polymersomes, emphasizing their ability to improve therapeutic outcomes through controlled drug release and targeted delivery. Moreover, we highlight the critical areas for future research, including the optimization of polymersome-biological interactions and biocompatibility, to facilitate their clinical adoption. Multi-responsive polymersomes emerge as a promising development in nanomedicine, offering a pathway to safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Irina Negut
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
| | - Bogdan Bita
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania;
- National Institute for Lasers, Plasma and Radiation Physics, 077125 Magurele, Romania
| |
Collapse
|
105
|
Adico MD, Bayala B, Zoure AA, Lagarde A, Bazie JT, Traore L, Buñay J, Yonli AT, Djigma F, Bambara HA, Baron S, Simporé J, Lobaccaro JMA. In vitro activities and mechanisms of action of anti-cancer molecules from African medicinal plants: a systematic review. Am J Cancer Res 2024; 14:1376-1401. [PMID: 38590420 PMCID: PMC10998760 DOI: 10.62347/auhb5811] [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: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 04/10/2024] Open
Abstract
Cancer is one of the leading causes of death worldwide. In recent years, African countries have been faced with a rapid increase in morbidity and mortality due to this pathology. Management is often complicated by the high treatment costs, side effects and the increasing occurrence of resistance to treatments. The identification of new active ingredients extracted from endemic medicinal plants is definitively an interesting approach for the implementation of new therapeutic strategies: their extraction is often lower cost; their identification is based on an ethnobotanical history and a tradipratic approach; their use by low-income populations is simpler; this can help in the development of new synthetic molecules that are more active, more effective and with fewer side effects. The objective of this review is to document the molecules derived from African medicinal plants whose in vitro anti-cancer activities and the mechanisms of molecular actions have been identified. From the scientific databases Science Direct, PubMed and Google Scholar, we searched for publications on compounds isolated from African medicinal plants and having activity on cancer cells in culture. The data were analyzed in particular with regard to the cytotoxicity of the compounds and their mode of action. A total of 90 compounds of these African medicinal plants were selected. They come from nine chemical groups: alkaloids, flavonoids, polyphenols, quinones, saponins, steroids, terpenoids, xanthones and organic sulfides. These compounds have been associated with several cellular effects: i) Cytotoxicity, including caspase activation, alteration of mitochondrial membrane potential, and/or induction of reactive oxygen species (ROS); ii) Anti-angiogenesis; iii) Anti-metastatic properties. This review points out that the cited African plants are rich in active ingredients with anticancer properties. It also stresses that screening of these anti-tumor active ingredients should be continued at the continental scale. Altogether, this work provides a rational basis for the selection of phytochemical compounds for use in clinical trials.
Collapse
Affiliation(s)
- Marc Dw Adico
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Bagora Bayala
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Ecole Normale Supérieure Koudougou, Burkina Faso
| | - Abdou A Zoure
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Laboratoire de recherches Biomédicales (LaReBio), Département de santé publique et biomédicale, Institut de Recherche en Sciences de la Santé (IRSS/CNRST) Ouagadougou, Burkina Faso
| | - Aurélie Lagarde
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Jean Tv Bazie
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Département des Substances Naturelles (DSN), Institut de Recherche en Sciences et Technologies Appliquées (IRSAT) Ouagadougou, Burkina Faso
| | - Lassina Traore
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Julio Buñay
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Albert T Yonli
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Florencia Djigma
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
| | - Hierrhum A Bambara
- Service d'oncologie, Centre hospitalier universitaire BOGODOGO, Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
| | - Silvère Baron
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| | - Jacques Simporé
- Laboratoire de Biologie Moléculaire et Génétique (LABIOGENE), Université Joseph KI-ZERBO Ouagadougou, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA) Ouagadougou, Burkina Faso
- Faculté de médecine, Université Saint Thomas d'Aquin (USTA) Ouagadougou, Burkina Faso
| | - Jean-Marc A Lobaccaro
- Institute Génétique, Reproduction, Développement, UMR CNRS 6293, INSERM U1103, Université Clermont Auvergne 28, Place Henri Dunant, BP38, F63001, Clermont-Ferrand, France
| |
Collapse
|
106
|
Kobyakova MI, Senotov AS, Krasnov KS, Lomovskaya YV, Odinokova IV, Kolotova AA, Ermakov AM, Zvyagina AI, Fadeeva IS, Fetisova EI, Akatov VS, Fadeev RS. Pro-Inflammatory Activation Suppresses TRAIL-induced Apoptosis of Acute Myeloid Leukemia Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:431-440. [PMID: 38648763 DOI: 10.1134/s0006297924030040] [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: 10/09/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 04/25/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is a promising agent for treatment of AML due to its specific apoptosis-inducing effect on tumor cells but not normal cells. However, emergence of resistance to TRAIL in the AML cells limits its potential as an antileukemic agent. Previously, we revealed increase in the resistance of the human AML THP-1 cells to the TRAIL-induced death during their LPS-dependent proinflammatory activation and in the in vitro model of LPS-independent proinflammatory activation - in a long-term high-density cell culture. In this study, we investigated mechanisms of this phenomenon using Western blot analysis, caspase 3 enzymatic activity analysis, quantitative reverse transcription-PCR, and flow cytometry. The results showed that the increased resistance to the TRAIL-induced cell death of AML THP-1 cells during their pro-inflammatory activation is associated with the decrease in the surface expression of the proapoptotic receptors TRAIL-R1/DR4 and TRAIL-R2/DR5, as well as with the increased content of members of the IAPs family - Livin and cIAP2. The results of this article open up new insights into the role of inflammation in formation of the resistance of AML cells to the action of mediators of antitumor immunity, in particular TRAIL.
Collapse
Affiliation(s)
- Margarita I Kobyakova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
- Institute of Clinical and Experimental Lymphology, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630060, Russia
| | - Anatoly S Senotov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Kirill S Krasnov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Yana V Lomovskaya
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Irina V Odinokova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anastasia A Kolotova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Artem M Ermakov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Alena I Zvyagina
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Irina S Fadeeva
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Elena I Fetisova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir S Akatov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
107
|
Goleij P, Sanaye PM, Babamohamadi M, Tabari MAK, Amirian R, Rezaee A, Mirzaei H, Kumar AP, Sethi G, Sadreddini S, Jeandet P, Khan H. Phytostilbenes in lymphoma: Focuses on the mechanistic and clinical prospects of resveratrol, pterostilbene, piceatannol, and pinosylvin. Leuk Res 2024; 138:107464. [PMID: 38422882 DOI: 10.1016/j.leukres.2024.107464] [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: 01/12/2024] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Lymphoma is a cancer affecting the lymphatic system that fights infections and diseases. In addition to surgery, radiotherapy, and chemotherapy, novel approaches have recently been investigated, such as phytostilbenes in treating lymphoma. Phytostilbenes are natural compounds present in various plants and have been shown to have different therapeutic effects, including anticancer properties. Resveratrol is a main phytostilbene with various derivates followed by pterostilbene and piceatannol. Studies have revealed that phytostilbenes can suppress the growth and proliferation of lymphoma cells by inducing apoptosis and inhibiting specific enzyme activity in cancer cell survival. The compounds also have antiinflammatory effects contributing to reducing lymphoma-associated inflammation. Additionally, phytostilbenes have been shown to increase the immune system's ability to fight cancer cells by activating immune cells (T-cells and natural killer cells). This review investigates the potential therapeutic effects of phytostilbenes, including resveratrol, pterostilbene, piceatannol, and pinosylvin, against lymphoma.
Collapse
Affiliation(s)
- Pouya Goleij
- Department of Genetics, Sana Institute of Higher Education, Sari, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Majma Sanaye
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehregan Babamohamadi
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran; Stem Cell and Regenerative Medicine Innovation Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran; USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roshanak Amirian
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sarvin Sadreddini
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Philippe Jeandet
- University of Reims Champagne-Ardenne, Research Unit "Induced Resistance and Plant Bioprotection", RIBP-USC INRA 1488, Reims 51100, France
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| |
Collapse
|
108
|
Alemi F, Poornajaf Y, Hosseini F, Vahedian V, Gharekhani M, Shoorei H, Taheri M. Interaction between lncRNAs and RNA-binding proteins (RBPs) influences DNA damage response in cancer chemoresistance. Mol Biol Rep 2024; 51:308. [PMID: 38366290 DOI: 10.1007/s11033-024-09288-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: 09/13/2023] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
The DNA damage response (DDR) is a crucial cellular signaling pathway activated in response to DNA damage, including damage caused by chemotherapy. Chemoresistance, which refers to the resistance of cancer cells to the effects of chemotherapy, poses a significant challenge in cancer treatment. Understanding the relationship between DDR and chemoresistance is vital for devising strategies to overcome this resistance and improve treatment outcomes. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that do not code for proteins but play important roles in various biological processes, including cancer development and chemoresistance. RNA-binding proteins (RBPs) are a group of proteins that bind to RNA molecules and regulate their functions. The interaction between lncRNAs and RBPs has been found to regulate gene expression at the post-transcriptional level, thereby influencing various cellular processes, including DDR signaling pathways. Multiple studies have demonstrated that lncRNAs can interact with RBPs to modulate the expression of genes involved in cancer chemoresistance by impacting DDR signaling pathways. Conversely, RBPs can regulate the expression and function of lncRNAs involved in DDR. Exploring these interactions can provide valuable insights for the development of innovative therapeutic approaches to overcome chemoresistance in cancer patients. This review article aims to summarize recent research on the interaction between lncRNAs and RBPs during cancer chemotherapy, with a specific focus on DDR pathways.
Collapse
Affiliation(s)
- Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Poornajaf
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Foroogh Hosseini
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Vahid Vahedian
- Department of Medical Clinic, Division of Hematology/Oncology and Cellular Therapy, Faculty of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Mahdi Gharekhani
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
- Rooyesh Infertility Center, Birjand University of Medical Sciences, Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
109
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Novel immunotherapies for breast cancer: Focus on 2023 findings. Int Immunopharmacol 2024; 128:111549. [PMID: 38266449 DOI: 10.1016/j.intimp.2024.111549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Immunotherapy has emerged as a revolutionary approach in cancer therapy, and recent advancements hold significant promise for breast cancer (BCa) management. Employing the patient's immune system to combat BCa has become a focal point in immunotherapeutic investigations. Strategies such as immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), and targeting the tumor microenvironment (TME) have disclosed encouraging clinical outcomes. ICIs, particularly programmed cell death protein 1 (PD-1)/PD-L1 inhibitors, exhibit efficacy in specific BCa subtypes, including triple-negative BCa (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive cancers. ACT approaches, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy, showed promising clinical outcomes in enhancing tumor recognition and elimination. Targeting the TME through immune agonists and oncolytic viruses signifies a burgeoning field of research. While challenges persist in patient selection, resistance mechanisms, and combination therapy optimization, these novel immunotherapies hold transformative potential for BCa treatment. Continued research and clinical trials are imperative to refine and implement these innovative approaches, paving the way for improved outcomes and revolutionizing the management of BCa. This review provides a concise overview of the latest immunotherapies (2023 studies) in BCa, highlighting their potential and current status.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Shi-Ya Yao
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Ke-Tao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
110
|
Perinbarajan GK, Sinclair BJ, Mossa AT, Ohja N, Jeelani PG. Silica/ Annona muricata nano-hybrid: Synthesis and anticancer activity against breast cancer. Heliyon 2024; 10:e25048. [PMID: 38322972 PMCID: PMC10844132 DOI: 10.1016/j.heliyon.2024.e25048] [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: 05/16/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/08/2024] Open
Abstract
Biogenically derived silica nanoparticles may serve as a well-defined target vehicle for drug delivery and have a wide range of applications in biomedicine. Silica nanoparticles are an excellent candidate as drug carriers due to their mesoporous structure, high drug loading capacity, low toxicity, environmental friendliness and low economic synthesis procedures. In this study, nano structured silica was extracted from sugarcane bagasse through an alkali leaching extraction and conjugated with A. muricata extract overcoming its poor solubility and improving its bioavailability within the host system. The Silica Nanoparticles (SNP) and Annona muricata conjugated Silica Nanoparticles (AM/SNP) were characterized using SEM, FTIR, TGA, EDAX, XRD and zeta potential. The AM/SNP was subjected to kinetic release studies and exhibited a sustained release of 64 % over the course of 12 h in contrast to extract, indicating the slow release of the drug under synthetic conditions. A. muricata pose a high affinity against tumor cells as an anti-cancer agent, and the potential of binding was testified using in-silico virtual screening against breast cancer receptors with lead acetogenins with Annomuricin (-7.4 kcal/mol) and Gigantecin (-7.4 kcal/mol) exhibiting a high binding affinity against ER and HER2+ receptors respectively. The AM/SNP conjugate exhibited high cytotoxicity against the MCF-7 breast cancer cell line with an IC50 value of 33.43 μg, indicating high potency of the conjugate at low concentrations, facilitating low systemic toxicity on administration.
Collapse
Affiliation(s)
- Gopi Krishna Perinbarajan
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, Tamil Nadu, India
| | - Bruce Joshua Sinclair
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, Tamil Nadu, India
| | - Abdel-Tawab Mossa
- National Research Centre, Egypt | Cairo, Egypt | NRC 33 El Buhouth St ‘Ad Doqi, Dokki, Cairo Governorate, 12622, Egypt
| | - Nupur Ohja
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, 600036, Tamil Nadu, India
| | - Peerzada Gh Jeelani
- Post Graduate and Research Department of Biotechnology & Microbiology National College (Autonomous), Tiruchirapalli, Tamilnadu, 620001, India
| |
Collapse
|
111
|
Abdallah R, Shaito AA, Badran A, Baydoun S, Sobeh M, Ouchari W, Sahri N, Eid AH, Mesmar JE, Baydoun E. Fractionation and phytochemical composition of an ethanolic extract of Ziziphus nummularia leaves: antioxidant and anticancerous properties in human triple negative breast cancer cells. Front Pharmacol 2024; 15:1331843. [PMID: 38405665 PMCID: PMC10885810 DOI: 10.3389/fphar.2024.1331843] [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: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 02/27/2024] Open
Abstract
Natural products have long been utilized in traditional medicine as remedies to improve health and treat illnesses, and have had a key role in modern drug discovery. Recently, there has been a revived interest in the search for bioactives from natural sources as alternative or complementary modalities to synthetic medicines; especially for cancer treatment, which incidence and mortality rates are on the rise worldwide. Ziziphus nummularia has been widely used in traditional medicine for the treatment of various diseases. Its traditional uses and numerous ethnopharmacological properties may be attributed to its richness in bioactive metabolites. However, its phytochemical composition or chemopreventive effects against the aggressive triple-negative breast cancer (TNBC) are still poorly explored. Here, phytochemical composition of an ethanolic extract of Z. nummularia leaves (ZNE) and its chromatographically isolated fractions was identified both qualitatively by spectrophotometric assays and analytically by HPLC-PDA-MS/MS. The anti-proliferative effects of ZNE were tested in several cancer cell lines, but we focused on its anti-TNBC effects since they were not explored yet. The anti-cancerous potential of ZNE and its fractions was tested in vitro in MDA-MB-231, a TNBC cell line. Results showed that ZNE and its Fraction 6 (F6) reduced the viability of MDA-MB-231 cells. F6 decreased MDA-MB-231 viability more than crude ZNE or its other fractions. ZNE and F6 are rich in phytochemicals and HPLC-PDA-MS/MS analysis identified several metabolites that were previously reported to have anti-cancerous effects. Both ZNE and F6 showed potent antioxidant capacity in the DPPH assay, but promoted reactive oxygen species (ROS) production in MDA-MB-231 cells; an effect which was blunted by the antioxidant N-acetyl cysteine (NAC). NAC also blunted ZNE- and F6-induced reduction in TNBC cell viability. We also demonstrated that ZNE and F6 induced an arrest of the cell cycle, and triggered apoptosis- and autophagy-mediated cell death. ZNE and F6 inhibited metastasis-related cellular processes by modifying cell migration, invasion, and adhesion. Taken together, our findings reveal that Z. nummularia is rich in phytochemicals that can attenuate the malignant phenotype of TNBC and may offer innovative avenues for the discovery of new drug leads for treatment of TNBC and other cancers.
Collapse
Affiliation(s)
- Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman, Jordan
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Mansour Sobeh
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Wafae Ouchari
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Nihad Sahri
- Agrobiosciences Program, College for Agriculture and Environmental Science, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
112
|
Pham DD, Pham TH, Bui TH, Britikova EV, Britikov VV, Bocharov EV, Usanov SA, Phan VC, Le TBT. In vitro and in vivo anti-tumor effect of Trichobakin fused with urokinase-type plasminogen activator ATF-TBK. Mol Biol Rep 2024; 51:130. [PMID: 38236367 DOI: 10.1007/s11033-023-09036-6] [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: 08/22/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Trichobakin (TBK), a member of type I ribosome-inactivating proteins (RIPs), was first successfully cloned from Trichosanthes sp Bac Kan 8-98 in Vietnam. Previous study has shown that TBK acts as a potential protein synthesis inhibitor; however, the inhibition efficiency and specificity of TBK on cancer cells remain to be fully elucidated. METHODS AND RESULTS In this work, we employed TBK and TBK conjugated with a part of the amino-terminal fragment (ATF) of the urokinase-type plasminogen activator (uPA), which contains the Ω-loop that primarily interacts with urokinase-type plasminogen activator receptor, and can be a powerful carrier in the drug delivery to cancer cells. Four different human tumor cell lines and BALB/c mice bearing Lewis lung carcinoma cells (LLC) were used to evaluate the role of TBK and ATF-TBK in the inhibition of tumor growth. Here we showed that the obtained ligand fused RIP (ATF-TBK) reduced the growth of four human cancer cell lines in vitro in the uPA receptor level-dependent manner, including the breast adenocarcinoma MDA-MB 231 cells and MCF7 cells, the prostate carcinoma LNCaP cells and the hepatocellular carcinoma HepG2 cells. Furthermore, the conjugate showed anti-tumor activity and prolonged the survival time of tumor-bearing mice. The ATF-TBK also did not cause the death of mice with doses up to 48 mg/kg, and they were not significantly distinct on parameters of hematology and serum biochemistry between the control and experiment groups. CONCLUSIONS In conclusion, ATF-TBK reduced the growth of four different human tumor cell lines and inhibited lung tumor growth in a mouse model with little side effects. Hence, the ATF-TBK may be a target to consider as an anti-cancer agent for clinical trials.
Collapse
Affiliation(s)
- Dan Duc Pham
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Hue Pham
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Huyen Bui
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Elena V Britikova
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Vladimir V Britikov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Eduard V Bocharov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997
| | - Sergey A Usanov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Van Chi Phan
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Bich Thao Le
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam.
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam.
| |
Collapse
|
113
|
Ahmadpour MR, Yousefi M, Rakhshandeh H, Darroudi M, Mousavi SH, Soukhtanloo M, Sabouri Z, Askari VR, Hashemzadeh A, Manjiri MA, Motavasselian M. Biosynthesis of Gold Nanoparticles Using Quince Seed Water Extract and Investigation of Their Anticancer Effect Against Cancer Cell Lines. IEEE Trans Nanobioscience 2024; 23:118-126. [PMID: 37379200 DOI: 10.1109/tnb.2023.3287805] [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: 06/30/2023]
Abstract
In this study, gold nanoparticles (Au-NPs) were synthesized using HAuCl4 and quince seed mucilage (QSM) extract, which was characterized by conventional methods including Fourier transforms electron microscopy (FTIR), UV-Visible spectroscopy (UV-Vis), Field emission electron microscopy (FESEM), Transmission electron microscopy (TEM), Dynamic light spectroscopy (DLS), and Zeta-potential. The QSM acted as reductant and stabilizing agents simultaneously. The NP's anticancer activity was also investigated against osteosarcoma cell lines (MG-63), which showed an IC50 of [Formula: see text]/mL.
Collapse
|
114
|
Zaer M, Moeinzadeh A, Abolhassani H, Rostami N, Tavakkoli Yaraki M, Seyedi SA, Nabipoorashrafi SA, Bashiri Z, Moeinabadi-Bidgoli K, Moradbeygi F, Farmani AR, Hossein-Khannazer N. Doxorubicin-loaded Niosomes functionalized with gelatine and alginate as pH-responsive drug delivery system: A 3D printing approach. Int J Biol Macromol 2023; 253:126808. [PMID: 37689301 DOI: 10.1016/j.ijbiomac.2023.126808] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Despite many efforts, breast cancer remains one of the deadliest cancers and its treatment faces challenges related to cancer drug side effects and metastasis. Combining 3D printing and nanocarriers has created new opportunities in cancer treatment. In this work, 3D-printed gelatin-alginate nanocomposites containing doxorubicin-loaded niosomes (Nio-DOX@GT-AL) were recruited as an advanced potential pH-sensitive drug delivery system. Morphology, degradation, drug release, flow cytometry, cell cytotoxicity, cell migration, caspase activity, and gene expression of nanocomposites and controls (Nio-DOX and Free-DOX) were evaluated. Results show that the obtained niosome has a spherical shape and size of 60-80 nm. Sustained drug release and biodegradability were presented by Nio-DOX@GT-AL and Nio-DOX. Cytotoxicity analysis revealed that the engineered Nio-DOX@GT-AL scaffold had 90 % cytotoxicity against breast cancer cells (MCF-7), whereas exhibited <5 % cytotoxicity against the non-tumor breast cell line (MCF-10A), which was significantly more than the antitumor effect of the control samples. Scratch-assay as an indicator cell migration demonstrated a reduction of almost 60 % of the covered surface. Gene expression could provide an explanation for the antitumor effect of engineered nanocarriers, which significantly reduced metastasis-promoting genes (Bcl2, MMP-2, and MMP-9), and significantly enhanced the expression and activity of genes that promote apoptosis (CASP-3, CASP-8, and CASP-9). Also, considerable inhibition of metastasis-associated genes (Bax and p53) was observed. Moreover, flow-cytometry data demonstrated that Nio-DOX@GT-AL decreased necrosis and enhanced apoptosis drastically. The findings of this research can confirm that employing 3D-printing and niosomal formulation can be an effective strategy in designing novel nanocarriers for efficient drug delivery applications.
Collapse
Affiliation(s)
- Mohammad Zaer
- Biomedical Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Alaa Moeinzadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Abolhassani
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Neda Rostami
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Seyed Arsalan Seyedi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran, Iran
| | - Seyed Ali Nabipoorashrafi
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kasra Moeinabadi-Bidgoli
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradbeygi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research, Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
115
|
Amiri S, Pashizeh F, Moeinabadi-Bidgoli K, Eyvazi Y, Akbari T, Salehi Moghaddam Z, Eskandarisani M, Farahmand F, Hafezi Y, Nouri Jevinani H, Seif M, Mousavi-Niri N, Chiani M, Tavakkoli Yaraki M. Co-encapsulation of hydrophilic and hydrophobic drugs into niosomal nanocarrier for enhanced breast cancer therapy: In silico and in vitro studies. ENVIRONMENTAL RESEARCH 2023; 239:117292. [PMID: 37806480 DOI: 10.1016/j.envres.2023.117292] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Combination therapy has been considered one of the most promising approaches for improving the therapeutic effects of anticancer drugs. This is the first study that uses two different antioxidants in full-characterized niosomal formulation and thoroughly evaluates their synergistic effects on breast cancer cells. In this study, in-silico studies of hydrophilic and hydrophobic drugs (ascorbic acid: Asc and curcumin: Cur) interactions and release were investigated and validated by a set of in vitro experiments to reveal the significant improvement in breast cancer therapy using a co-delivery approach by niosomal nanocarrier. The niosomal nanoparticles containing surfactants (Span 60 and Tween 60) and cholesterol at 2:1 M ratio were prepared through the film hydration method. A systematic evaluation of nanoniosomes was carried out. The release profile demonstrated two phases (initial burst followed by sustained release) and a pH-dependent release schedule over 72 h. The optimized niosomal preparation displayed superior storage stability for up to 2 months at 4 °C, exhibiting extremely minor changes in pharmaceutical encapsulation efficiency and size. Free dual drugs (Asc + Cur) and dual-drug loaded niosomes (Niosomal (Asc + Cur)) enhanced the apoptotic activity and cytotoxicity and inhibited cell migration which confirmed the synergistic effect of co-encapsulated drugs. Also, significant up-regulation of p53 and Bax genes was observed in cells treated with Asc + Cur and Niosomal (Asc + Cur), while the anti-apoptotic Bcl-2 gene was down-regulated. These results were in correlation with the increase in the enzyme activity of SOD, CAT, and caspase, and the levels of malondialdehyde (MDA) and reactive oxygen species (ROS) upon treatment with the mentioned drugs. Furthermore, these anti-cancer effects were higher when using Niosomal (Asc + Cur) than Asc + Cur. Histopathological examination also revealed that Niosomal (Asc + Cur) had a lower mitosis index, invasion, and pleomorphism than Asc + Cur. These findings indicated that niosomal formulation for co-delivery of Asc and Cur would offer a promising delivery system for an effective breast cancer treatment.
Collapse
Affiliation(s)
- Sahar Amiri
- Department of Genetic, Islamic Azad University, Tehran North Branch, Iran
| | - Fatemeh Pashizeh
- Department of Immunology, School of Medicine, Shahid Sadoughi University of Medical Science Yazd, Iran
| | - Kasra Moeinabadi-Bidgoli
- Departments of Medicine and Endocrinology, University of California San Francisco and San Francisco Veterans Affairs Health Center, San Francisco, CA, USA
| | - Yalda Eyvazi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Tanin Akbari
- Department of Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi Moghaddam
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, Iran
| | | | - Faranak Farahmand
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Hafezi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hoda Nouri Jevinani
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdi Seif
- Faculty of Materials Science and Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | - Neda Mousavi-Niri
- Department of Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohsen Chiani
- Department of Nano Biotechnology, New Technology Research Group, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, NSW 2109, Australia.
| |
Collapse
|
116
|
Kadarullah O, Tamtomo DG, Wasita B, Setiamika M. Animal Model of Cisplatin-Induced Oral Mucositis: Dose Optimization. Acta Med Acad 2023; 52:188-194. [PMID: 38407085 PMCID: PMC10945320 DOI: 10.5644/ama2006-124.422] [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: 07/20/2023] [Accepted: 10/24/2023] [Indexed: 02/27/2024] Open
Abstract
OBJECTIVE The present study aimed to develop and validate an animal model of chemotherapy-induced oral mucositis due to cisplatin administration. MATERIALS AND METHODS Oral mucositis was induced in Wistar rats by cisplatin. Twenty healthy male Wistar rats were divided into four groups: a control group, and cisplatin 3 mg/kgBW (D1), cisplatin 5 mg/kgBW (D2), and cisplatin 6 mg/kgBW groups (D3). The D1, D2, and D3 groups received the cisplatin intraperitoneally on days 1, 3, and 5, whereas the control group did not receive anything. On day 7 and day 14 the entire experiment was terminated in all groups and the changes in body weight, oral mucositis grades, and histopathological scores were evaluated. RESULTS Cisplatin administration created a strong oral mucositis effect on groups D2 and D3. All the cisplatin doses decreased the rats' body weight by day 14. The worst oral mucositis grades and histopathological scores resulted from the administration of cisplatin at a dose of 5 mg/kgBW. CONCLUSIONS In conclusion the cisplatin 5 mg/kgBW administered on days 1, 3, and 5 by intraperitoneal administration was the optimum dose to induce oral mucositis.
Collapse
Affiliation(s)
- Oke Kadarullah
- Doctoral Program of Medical Sciences, Faculty of Medicine, University of Sebelas Maret, Surakarta, Central Java, Indonesia; Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, University of Muhammadiyah Purwokerto, Banyumas, Central Java, Indonesia. ; ORCID ID: 0000-0002-5237-2063
| | - Didik Gunawan Tamtomo
- Doctoral Program of Medical Sciences, Faculty of Medicine, University of Sebelas Maret, Surakarta, Central Java, Indonesia
| | - Brian Wasita
- Department of Anatomical Pathology, Faculty of Medicine, University of Sebelas Maret, Surakarta, Central Java, Indonesia
| | - Made Setiamika
- Department of Otorhinolaryngology Head and Neck Surgery, Dr. Moewardi Hospital, Surakarta, Central Java, Indonesia
| |
Collapse
|
117
|
Luiz-Ferreira A, Pacifico T, Cruz ÁC, Laudisi F, Monteleone G, Stolfi C. TRAIL-Sensitizing Effects of Flavonoids in Cancer. Int J Mol Sci 2023; 24:16596. [PMID: 38068921 PMCID: PMC10706592 DOI: 10.3390/ijms242316596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
Collapse
Affiliation(s)
- Anderson Luiz-Ferreira
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Álefe Cardoso Cruz
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| |
Collapse
|
118
|
Cunha A, Silva PMA, Sarmento B, Queirós O. Targeting Glucose Metabolism in Cancer Cells as an Approach to Overcoming Drug Resistance. Pharmaceutics 2023; 15:2610. [PMID: 38004589 PMCID: PMC10675572 DOI: 10.3390/pharmaceutics15112610] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/27/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
The "Warburg effect" consists of a metabolic shift in energy production from oxidative phosphorylation to glycolysis. The continuous activation of glycolysis in cancer cells causes rapid energy production and an increase in lactate, leading to the acidification of the tumour microenvironment, chemo- and radioresistance, as well as poor patient survival. Nevertheless, the mitochondrial metabolism can be also involved in aggressive cancer characteristics. The metabolic differences between cancer and normal tissues can be considered the Achilles heel of cancer, offering a strategy for new therapies. One of the main causes of treatment resistance consists of the increased expression of efflux pumps, and multidrug resistance (MDR) proteins, which are able to export chemotherapeutics out of the cell. Cells expressing MDR proteins require ATP to mediate the efflux of their drug substrates. Thus, inhibition of the main energy-producing pathways in cancer cells, not only induces cancer cell death per se, but also overcomes multidrug resistance. Given that most anticancer drugs do not have the ability to distinguish normal cells from cancer cells, a number of drug delivery systems have been developed. These nanodrug delivery systems provide flexible and effective methods to overcome MDR by facilitating cellular uptake, increasing drug accumulation, reducing drug efflux, improving targeted drug delivery, co-administering synergistic agents, and increasing the half-life of drugs in circulation.
Collapse
Affiliation(s)
- Andrea Cunha
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences—CESPU (IUCS—CESPU), 4585-116 Gandra, Portugal; (A.C.); (P.M.A.S.); (B.S.)
| | - Patrícia M. A. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences—CESPU (IUCS—CESPU), 4585-116 Gandra, Portugal; (A.C.); (P.M.A.S.); (B.S.)
- 1H—TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences—CESPU (IUCS—CESPU), 3810-193 Gandra, Portugal
| | - Bruno Sarmento
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences—CESPU (IUCS—CESPU), 4585-116 Gandra, Portugal; (A.C.); (P.M.A.S.); (B.S.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Odília Queirós
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences—CESPU (IUCS—CESPU), 4585-116 Gandra, Portugal; (A.C.); (P.M.A.S.); (B.S.)
| |
Collapse
|
119
|
Khan S, Falahati M, Cho WC, Vahdani Y, Siddique R, Sharifi M, Jaragh-Alhadad LA, Haghighat S, Zhang X, Ten Hagen TLM, Bai Q. Core-shell inorganic NP@MOF nanostructures for targeted drug delivery and multimodal imaging-guided combination tumor treatment. Adv Colloid Interface Sci 2023; 321:103007. [PMID: 37812992 DOI: 10.1016/j.cis.2023.103007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 08/16/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
It is well known that metal-organic framework (MOF) nanostructures have unique characteristics such as high porosity, large surface areas and adjustable functionalities, so they are ideal candidates for developing drug delivery systems (DDSs) as well as theranostic platforms in cancer treatment. Despite the large number of MOF nanostructures that have been discovered, conventional MOF-derived nanosystems only have a single biofunctional MOF source with poor colloidal stability. Accordingly, developing core-shell MOF nanostructures with good colloidal stability is a useful method for generating efficient drug delivery, multimodal imaging and synergistic therapeutic systems. The preparation of core-shell MOF nanostructures has been done with a variety of materials, but inorganic nanoparticles (NPs) are highly effective for drug delivery and imaging-guided tumor treatment. Herein, we aimed to overview the synthesis of core-shell inorganic NP@MOF nanostructures followed by the application of core-shell MOFs derived from magnetic, quantum dots (QDs), gold (Au), and gadolinium (Gd) NPs in drug delivery and imaging-guided tumor treatment. Afterward, we surveyed different factors affecting prolonged drug delivery and cancer therapy, cellular uptake, biocompatibility, biodegradability, and enhanced permeation and retention (EPR) effect of core-shell MOFs. Last but not least, we discussed the challenges and the prospects of the field. We envision this article may hold great promise in providing valuable insights regarding the application of hybrid nanostructures as promising and potential candidates for multimodal imaging-guided combination cancer therapy.
Collapse
Affiliation(s)
- Suliman Khan
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| | - Yasaman Vahdani
- Department of Biochemistry and Molecular Medicine, University of Montreal, Canada
| | - Rabeea Siddique
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, the Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC, Rotterdam, the Netherlands.
| | - Qian Bai
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
120
|
Klemt I, Varzatskii O, Selin R, Vakarov S, Kovalska V, Bila G, Bilyy R, Voloshin Y, Cuartero IC, Hidalgo A, Frey B, Becker I, Friedrich B, Tietze R, Friedrich RP, Alexiou C, Ursu EL, Rotaru A, Solymosi I, Pérez-Ojeda ME, Mokhir A. 3D-Shaped Binders of Unfolded Proteins Inducing Cancer Cell-Specific Endoplasmic Reticulum Stress In Vitro and In Vivo. J Am Chem Soc 2023; 145:22252-22264. [PMID: 37773090 DOI: 10.1021/jacs.3c08827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
The amount of unfolded proteins is increased in cancer cells, leading to endoplasmic reticulum (ER) stress. Therefore, cancer cells are sensitive to drugs capable of further enhancing ER stress. Examples of such drugs include the clinically approved proteosome inhibitors bortezomib and carfilzomib. Unfortunately, the known ER stress inducers exhibit dose-limiting side effects that justify the search for better, more cancer-specific drugs of this type. Herein, we report on FeC 2, which binds to unfolded proteins prevents their further processing, thereby leading to ER stress and ROS increase in cancer cells, but not in normal cells. FeC 2 exhibits low micromolar toxicity toward human acute promyelocytic leukemia HL-60, Burkitt's lymphoma BL-2, T-cell leukemia Jurkat, ovarian carcinoma A2780, lung cancer SK-MES-1, and murine lung cancer LLC1 cells. Due to the cancer-specific mode of action, 2 is not toxic in vivo up to the dose of 147 mg/kg, does not affect normal blood and bone marrow cells at the therapeutically active dose, but strongly suppresses both primary tumor growth (confirmed in Nemeth-Kellner lymphoma and LLC1 lung cancer models of murine tumor) and spreading of metastases (LLC1).
Collapse
Affiliation(s)
- Insa Klemt
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Oleg Varzatskii
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- V.I. Vernadsky Institute of General and Inorganic Chemistry, NASU, 32/34 Palladin Av., 03142 Kyiv, Ukraine
| | - Roman Selin
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Serhii Vakarov
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- V.I. Vernadsky Institute of General and Inorganic Chemistry, NASU, 32/34 Palladin Av., 03142 Kyiv, Ukraine
| | - Vladyslava Kovalska
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- Institute of Molecular Biology and Genetics, NASU, 150 Zabolotnogo Street, 03143 Kyiv, Ukraine
| | - Galyna Bila
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, Pekarska Street 69, 79010 Lviv, Ukraine
- Lectinotest R&D, Mechanichna Street 2, 79024 Lviv, Ukraine
| | - Rostyslav Bilyy
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, Pekarska Street 69, 79010 Lviv, Ukraine
- Lectinotest R&D, Mechanichna Street 2, 79024 Lviv, Ukraine
| | - Yan Voloshin
- Nesmeyanov Institute of Organoelement Compounds, RAS, 28 Vavilova Street, 119334 Moscow, Russia
| | - Itziar Cossío Cuartero
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), C. Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Andrés Hidalgo
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), C. Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Benjamin Frey
- Department of Radiation Oncology, Translational Radiobiology, Universitaetsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glueckstrasse 4A, 91054 Erlangen, Germany
| | - Ina Becker
- Department of Radiation Oncology, Translational Radiobiology, Universitaetsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glueckstrasse 4A, 91054 Erlangen, Germany
| | - Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Ralf P Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Elena-Laura Ursu
- "Petru Poni" Institute of Macromolecular Chemistry, Romanian Academy, Centre of Advanced Research in Bionanoconjugates and Biopolymers, Grigore Ghica Voda Alley 41 A, 700487 Iasi, Romania
| | - Alexandru Rotaru
- "Petru Poni" Institute of Macromolecular Chemistry, Romanian Academy, Centre of Advanced Research in Bionanoconjugates and Biopolymers, Grigore Ghica Voda Alley 41 A, 700487 Iasi, Romania
| | - Iris Solymosi
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - M Eugenia Pérez-Ojeda
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Andriy Mokhir
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| |
Collapse
|
121
|
Mollaei M, Homayouni Tabrizi M, Es-Haghi A. The folate-linked chitosan-coated Kaempferol/HSA nano-transporters (FCKH-NTs) as the selective apoptotic inducer in human MCF-7 breast cancer cell line. Drug Dev Ind Pharm 2023; 49:658-665. [PMID: 37814890 DOI: 10.1080/03639045.2023.2268739] [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: 04/19/2023] [Accepted: 10/04/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Kaempferol, the natural bioactive flavonoid, has been utilized as an efficient anti-breast cancer compound. In the current study, the Kaempferol's cellular uptake and its aqueous solubility were improved by using human serum albumin (HSA) as the Kaempferol adjuvant and encapsulating it with the folate-linked chitosan polymer to evaluate the apoptotic, activity of the novel-formulated Kaempferol in human MCF-7 breast cancer cells. METHODS The folate-linked chitosan-coated Kaempferol/HSA nano-transporters (FCKH-NTs) were synthesized and characterized using FTIR, FESEM, DLS, and Zeta potential analysis. The nano-transporters' selective cytotoxicity was studied by applying an MTT assay on the cancerous MCF-7 cells compared with normal HFF cell lines. Cell death type determination was determined by analyzing the expression of apoptotic (BAX and Cas-8) and anti-apoptotic genes (BCL2 and NF-κB). The FCKH-NTs apoptotic activity was verified by studying the flow cytometry and AO/PI staining results. RESULT The 126-nm FCKH-NTs (PDI = 0.282) selectively induced apoptotic death in human MCF-7 breast cancer cells by up-regulating the BAX, Nf- κB, and Cas-8 gene expression. The apoptotic activity of FCKH-NTs was verified by detecting the SubG1-arrested cancer cells and increased apoptotic bodies in AO/PI staining images. CONCLUSION The FCKH-NTs exhibited a selective-cytotoxic impact on human MCF-7 breast cancer cells compared with normal HFF cells, which can be due to the folate receptor-mediated endocytosis mechanism of the nano-transporters. Therefore, the FCKH-NTs have the potential to be used as a selective anti-breast cancer compound.
Collapse
Affiliation(s)
- Mahshad Mollaei
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Ali Es-Haghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
122
|
Rashidzadeh H, Ramazani A, Tabatabaei Rezaei SJ, Danafar H, Rahmani S, Veisi H, Rajaeinejad M, Jamalpoor Z, Hami Z. Targeted co-delivery of methotrexate and chloroquine via a pH/enzyme-responsive biocompatible polymeric nanohydrogel for colorectal cancer treatment. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1824-1842. [PMID: 36869798 DOI: 10.1080/09205063.2023.2187986] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/05/2023]
Abstract
Application of conventional chemotherapy regardless of its unique effectiveness have been gradually being edged aside due to limited targeting capability, lack of selectivity and chemotherapy-associated side effects. To this end, colon-targeted nanoparticles via combination therapy have shown great therapeutic potential against cancer. Herein, pH/enzyme-responsive biocompatible polymeric nanohydrogels based on poly(methacrylic acid) (PMAA) containing methotrexate (MTX) and chloroquine (CQ) were fabricated. PMAA-MTX-CQ exhibited high drug loading capacity of which MTX was 4.99% and was CQ 25.01% and displayed pH/enzyme-triggered drug release behavior. Higher CQ release rate (76%) under simulated acidic microenvironment of tumor tissue whereas 39% of CQ was released under normal physiological conditions. Intestinally, MTX release was facilitated in the presence of proteinase K enzyme. TEM image demonstrated spherical morphology with particle size of less than 50 nm. In vitro and in vivo toxicity assessments indicated that developed nanoplatforms possessed great biocompatibility. These nanohydrogels did not cause any adverse effects against Artemia Salina and HFF2 cells (around 100% cell viability) which highlight the safety of prepared nanohydrogels. There was no death in mice received different concentrations of nanohydrogel through oral administration and less than 5% hemolysis was found in red blood cells incubated with PMAA nanohydrogels. In vitro anti-cancer results showed that combination therapy based on PMAA-MTX-CQ can effectively suppress the growth of SW480 colon cancer cells (29% cell viability) compared to monotherapy. Altogether, these findings suggest that pH/enzyme-responsive PMAA-MTX-CQ could effectively inhibit cancer cell growth and progression via site-specific delivery of its cargo in a safe and controlled manner.
Collapse
Affiliation(s)
- Hamid Rashidzadeh
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Jamal Tabatabaei Rezaei
- Laboratory of Novel Drug Delivery Systems, Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shayan Rahmani
- Laboratory of Novel Drug Delivery Systems, Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, Iran
| | - Hassan Veisi
- Laboratory of Novel Drug Delivery Systems, Department of Chemistry, Faculty of Science, University of Zanjan, Zanjan, Iran
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Zahra Hami
- Toxicology Research Center, Aja University of Medical Sciences, Tehran, Iran
| |
Collapse
|
123
|
Terry S, Gommet C, Kerangueven AC, Leguet M, Thévenin V, Berthelot M, Begoud L, Windenberger F, Lainee P. Activity in Group-Housed Home Cages of Mice as a Novel Preclinical Biomarker in Oncology Studies. Cancers (Basel) 2023; 15:4798. [PMID: 37835492 PMCID: PMC10571829 DOI: 10.3390/cancers15194798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Improving experimental conditions in preclinical animal research is a major challenge, both scientifically and ethically. Automated digital ventilated cages (DVC®) offer the advantage of continuous monitoring of animal activity in their home-cage. The potential utility of this technology remains understudied and deserves investigation in the field of oncology. METHODS Using the DVC® platform, we sought to determine if the continuous assessment of locomotor activity of mice in their home cages can serve as useful digital readout in the monitoring of animals treated with the reference oncology compounds cisplatin and cyclophosphamide. SCID mice of 14 weeks of age were housed in DVC® cages in groups of four and followed with standard and digital examination before and after treatment over a 17-day total period. RESULTS DVC® detected statistically significant effects of cisplatin on the activity of mice in the short and long term, as well as trends for cyclophosphamide. The activity differences between the vehicle- and chemotherapy-treated groups were especially marked during the nighttime, a period when animals are most active and staff are generally not available for regular checks. Standard clinical parameters, such as body weight change and clinical assessment during the day, provided additional and complementary information. CONCLUSION The DVC® technology enabled the home cage monitoring of mice and non-invasive detection of animal activity disturbances. It can easily be integrated into a multimodal monitoring approach to better capture the different effects of oncology drugs on anti-tumor efficacy, toxicity, and safety and improve translation to clinical studies.
Collapse
Affiliation(s)
| | - Céline Gommet
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Anne-Cécile Kerangueven
- Biostatistics & Programming, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (A.-C.K.); (F.W.)
| | - Mickaël Leguet
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Vincent Thévenin
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Mickaël Berthelot
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Laurent Begoud
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| | - Fanny Windenberger
- Biostatistics & Programming, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (A.-C.K.); (F.W.)
| | - Pierre Lainee
- Translational In Vivo Models—In Vivo Research Center Vitry, Sanofi Research and Development, 94403 Vitry-sur-Seine, France; (C.G.); (M.L.); (V.T.); (M.B.); (L.B.)
| |
Collapse
|
124
|
Zhou Z, Zhang Y, Xia S, Chen X. Red-Light-Activatable AND-Gated Antitumor Immunosuppressant. Cells 2023; 12:2351. [PMID: 37830565 PMCID: PMC10571834 DOI: 10.3390/cells12192351] [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: 07/12/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023] Open
Abstract
Immunosuppressants are emerging as promising candidates for cancer therapy with lower cytotoxicity compared to traditional chemotherapy drugs; yet, the intrinsic side effects such as immunosuppression remain a critical concern. Herein, we introduce a photoactivatable antitumor immunosuppressant called dmBODIPY-FTY720 (BF) that shows no cytotoxicity but can be temporally and locally activated by deep-red light illumination to induce tumor cell apoptosis. To further reduce potential side effects, we integrate BF with another classic photosensitizer called methylene blue (MB) that is activated under the same wavelength of deep-red light (>650 nm) and successfully establish a red-light-activatable AND Boolean logic gate through a mechanism that we found to be synergetic apoptotic induction. At further decreased dosages, deep-red light illumination does not induce cell death in the presence of either BF or MB, but significant cancer cell death is triggered in the presence of both drugs. Therefore, the dosage of BF is further reduced, which will be highly beneficial to minimize any potential side effects of BF. This AND-gated strategy has been successfully applied in vivo for effective suppression of hepatocarcinoma tumors in living mice.
Collapse
Affiliation(s)
- Ziqi Zhou
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| | - Yan Zhang
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| | - Simin Xia
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
| | - Xi Chen
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| |
Collapse
|
125
|
Gatto MS, Najahi-Missaoui W. Lyophilization of Nanoparticles, Does It Really Work? Overview of the Current Status and Challenges. Int J Mol Sci 2023; 24:14041. [PMID: 37762348 PMCID: PMC10530935 DOI: 10.3390/ijms241814041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Nanoparticles are being increasingly used as drug delivery systems to enhance the delivery to and uptake by target cells and to reduce off-target toxicity of free drugs. However, although the advantages of nanoparticles as drug carriers are clear, there are still some limitations, especially in maintaining their long-term stability. Lyophilization, also known as freeze-drying, has been heavily investigated as a solution to this problem. This strategy has been shown to be effective in increasing both the long-term stability of nanoparticles and the shelf life of the drug product. However, the process is still in need of improvement in several aspects, such as the process parameters, formulation factors, and characterization techniques. This review summarizes the advantages and limitations of nanoparticles for the treatment of disease, advantages and limitations, and the status of the lyophilization of nanoparticles for therapeutic use and provides insight into both the advantages and the limitations.
Collapse
Affiliation(s)
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
126
|
Mapuskar KA, Pulliam CF, Zepeda-Orozco D, Griffin BR, Furqan M, Spitz DR, Allen BG. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2023; 12:1728. [PMID: 37760031 PMCID: PMC10525889 DOI: 10.3390/antiox12091728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin, a potent chemotherapeutic agent, is marred by severe nephrotoxicity that is governed by mechanisms involving oxidative stress, inflammation, and apoptosis pathways. The transcription factor Nrf2, pivotal in cellular defense against oxidative stress and inflammation, is the master regulator of the antioxidant response, upregulating antioxidants and cytoprotective genes under oxidative stress. This review discusses the mechanisms underlying chemotherapy-induced kidney injury, focusing on the role of Nrf2 in cancer therapy and its redox regulation in cisplatin-induced kidney injury. We also explore Nrf2's signaling pathways, post-translational modifications, and its involvement in autophagy, as well as examine redox-based strategies for modulating Nrf2 in cisplatin-induced kidney injury while considering the limitations and potential off-target effects of Nrf2 modulation. Understanding the redox regulation of Nrf2 in cisplatin-induced kidney injury holds significant promise for developing novel therapeutic interventions. This knowledge could provide valuable insights into potential strategies for mitigating the nephrotoxicity associated with cisplatin, ultimately enhancing the safety and efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Diana Zepeda-Orozco
- Pediatric Nephrology and Hypertension at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin R. Griffin
- Division of Nephrology, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
127
|
Muchtaridi M, Suryani AI, Wathoni N, Herdiana Y, Mohammed AFA, Gazzali AM, Lesmana R, Joni IM. Chitosan/Alginate Polymeric Nanoparticle-Loaded α-Mangostin: Characterization, Cytotoxicity, and In Vivo Evaluation against Breast Cancer Cells. Polymers (Basel) 2023; 15:3658. [PMID: 37765512 PMCID: PMC10538075 DOI: 10.3390/polym15183658] [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: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
α-mangostin (Amg), a compound isolated from the mangosteen rind (Garcinia mangostana, L.), has demonstrated promising anticancer activity. However, its low solubility and selectivity against cancer cells limit its efficacy. To address this issue, researchers have developed chitosan/alginate polymeric nanoparticles (NANO-AMCAL) to enhance the effectiveness of Amg. In vitro studies have demonstrated that NANO-AMCAL is highly active against breast cancer cells. Therefore, an in vivo study was conducted to evaluate the efficacy of NANO-AMCAL in treating breast cancer in Wistar rats (Rattus norvegicus) and determine the effective dose. The rats were divided into seven treatment groups, including positive control, negative control, pure Amg, and NANO-AMCAL 5 mg, 10 mg, and 20 mg. The rats were injected subcutaneously with a carcinogenic agent, 7,12-dimethylbenz(a)anthracene (DMBA) and were evaluated for weight and tumor volume every three days during treatment. Surgery was performed on day 14, and histopathological studies were carried out on breast and lung cancer tissues. The results showed that NANO-AMCAL significantly enhanced the anticancer activity of Amg in treating breast cancer in Wistar rats. NANO-AMCAL containing 0.33 mg of Amg had a healing effect three times better than 20 mg pure Amg and was comparable to tamoxifen. The effective dose of NANO-AMCAL for anti-breast cancer treatment in Wistar rats was found to be 20 mg, which exhibited a good healing response, and the tumor volume continued to decrease up to 17.43% on the 14th day. Furthermore, histopathological tests showed tissue repair and no metastases. These findings suggest that NANO-AMCAL may be a promising therapeutic option for breast cancer treatment.
Collapse
Affiliation(s)
- Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jalan Raya Bandung-Sumedang Km 21, Jatinangor 45363, Indonesia
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Jakarta 10340, Indonesia
| | - Ade Irma Suryani
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (N.W.); (Y.H.)
| | - Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (N.W.); (Y.H.)
| | | | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Penang, Malaysia;
| | - Ronny Lesmana
- Physiology Division, Department of Anatomy, Physiology and Biology Cell, Faculty of Medicine, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - I. Made Joni
- Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jalan Raya Bandung-Sumedang Km 21, Jatinangor 45363, Indonesia
- Departement of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
128
|
Elfiky AA, Ibrahim IM, Elghareib AM, Bashandy YS, Samir A, Hamdy MM, Kamal RT, Amin FG, Elkaramany Y, Rashad AM, Abdelaziz YS, Fathey MM. Simulation of gold nanoparticle movement through normal and cancer cell membranes. Comput Biol Med 2023; 164:107363. [PMID: 37595520 DOI: 10.1016/j.compbiomed.2023.107363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/18/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023]
Abstract
Gold nanoparticles (Au-NPs) have been used for a long time to target cancer cells. Different modalities have been suggested to utilize Au-NPs in cancer patients. We construct both normal and cancer cell membranes to simulate the Au-NP entry to understand better how it can penetrate the cancer cell membrane. We use molecular dynamics simulation (MDS) on both normal and cancer cell membrane models for 150 ns. At the same time, we prepared the Au-NP of spherical shape (16 nm radius) capped with citrate using MDS for 100 ns. Finally, we added the Au-NP close to the membranes and moved it using 1000 kJ mol-1 nm-1 force constant during the 7.7 ns MDS run. We analyzed the membranes in the presence and absence of the Au-NP and compared normal and cancer membranes. The results show that normal cell membranes have higher stability than cancer membranes. Additionally, Au-NP forms pore in the membranes that facilitate water and ions entry during the movement inside the lipid bilayer region. These pores are responsible for the enhanced response of Au-NP-loaded chemotherapeutic agents in cancer treatment.
Collapse
Affiliation(s)
- Abdo A Elfiky
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt.
| | - Ibrahim M Ibrahim
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed M Elghareib
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Yousef S Bashandy
- Biotechnology and Biomolecular Chemistry Department, Cairo University, Giza, Egypt
| | - Ahmed Samir
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Mayar M Hamdy
- Biotechnology and Biomolecular Chemistry Department, Cairo University, Giza, Egypt
| | - Rana T Kamal
- Biotechnology and Biomolecular Chemistry Department, Cairo University, Giza, Egypt
| | - Fatma G Amin
- Physics Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Yomna Elkaramany
- Biotechnology and Biomolecular Chemistry Department, Cairo University, Giza, Egypt
| | - Anan M Rashad
- Biotechnology and Biomolecular Chemistry Department, Cairo University, Giza, Egypt
| | - Youssef S Abdelaziz
- Department of Potany and Microbiology, Faculty of Science, Cairo University, Giza, Egypt
| | - Mohamed M Fathey
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
129
|
GÜRAĞAÇ DERELİ FT, AKKOÇ S. Investigation of In Vitro antiproliferative activity properties of Spartium junceum L. (Spanish broom) against MDA-MB-231 and HepG2 cancer cell lines. INTERNATIONAL JOURNAL OF SECONDARY METABOLITE 2023; 10:345-353. [DOI: 10.21448/ijsm.1182965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Cancer is among the top global public health burdens leading to millions of deaths each year. The study aims to investigate the antiproliferative effect of Spartium junceum L. flowers on different cancer cell lines. The ethanolic extract of the flowers was prepared in the present study. Phytochemical analysis of the plant extract revealed the presence of several phenolic compounds such as cinnamic acid and its derivatives (chlorogenic, p-coumaric, ferulic acids), protocatechuic acid, epicatechin and luteolin. This extract was tested against human breast (MDA-MB-231) and liver (HepG2) cancer cell lines to find out its antiproliferative activity. It was determined that the extract was effective against both cell lines with IC50 values of 2.37 ± 0.47 and 0.98 ± 0.01 µL/mL for MDA-MB-231 and HepG2, respectively. Particularly, the extract was found to be more effective in the liver cancer cell line than the breast cancer cell line. All these obtained findings led us to believe that this medicinal plant could be a promising antiproliferative agent candidate for the treatment of human liver and breast cancers.
Collapse
|
130
|
Westwood L, Emmerson E, Callanan A. Fabrication of polycaprolactone electrospun fibres with retinyl acetate for antioxidant delivery in a ROS-mimicking environment. Front Bioeng Biotechnol 2023; 11:1233801. [PMID: 37650040 PMCID: PMC10463743 DOI: 10.3389/fbioe.2023.1233801] [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: 06/02/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background: Increased cancer rates denote that one in two people will be diagnosed with cancer in their lifetime. Over 60% of cancer patients receive radiotherapy, either as a stand-alone treatment or in combination with other treatments such as chemotherapy and surgery. Whilst radiotherapy is effective in destroying cancer cells, it also causes subsequent damage to healthy cells and surrounding tissue due to alterations in the tumor microenvironment and an increase in reactive oxygen species (ROS). This can cause extensive damage that impairs tissue function, and the likelihood of tissue regeneration and restoration of function is significantly reduced as new healthy cells cannot survive in the damaged environment. In the treatment of head and neck cancers, radiotherapy can cause salivary gland dysfunction. This significantly impairs the patient's quality of life and there is currently no cure, only palliative treatment options. Tissue engineering approaches are used to mimic the microenvironment of the tissue and can mediate the damaged microenvironment via bioactive compounds, to support the delivery, survival, and proliferation of new, healthy cells into the damaged environment. Methods: In this study, retinyl acetate, a derivative of vitamin A, was successfully incorporated into electrospun polycaprolactone fibres. Results: SEM images and characterization analyses showed that all scaffolds produced had similar characteristics, including fiber morphology and scaffold wettability. The vitamin scaffolds were shown to exert an antioxidant effect through scavenging activity of both DPPH and hydroxyl radicals in vitro. Critically, the antioxidant scaffolds supported the growth of human submandibular gland cells and significantly upregulated the expression of GPx1, an antioxidant enzyme, when cultured under both normal conditions and under a simulated oxidative stress environment. Discussion: These results suggest that incorporation of retinyl acetate into electrospun fibres has may mediate the damaged microenvironment post cancer radiation therapy.
Collapse
Affiliation(s)
- Lorna Westwood
- School of Engineering, Institute for Bioengineering, University of Edinburgh, Edinburgh, United Kingdom
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine Emmerson
- The Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Anthony Callanan
- School of Engineering, Institute for Bioengineering, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
131
|
Cesnik R, Toohey K, Freene N, Kunstler B, Semple S. Physical Activity Levels in People with Cancer Undergoing Chemotherapy: A Systematic Review. Semin Oncol Nurs 2023; 39:151435. [PMID: 37127520 DOI: 10.1016/j.soncn.2023.151435] [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: 12/21/2022] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 05/03/2023]
Abstract
OBJECTIVES Physical activity (PA) has been shown to improve chemotherapy side effects, survival rates and treatment adherence in people with cancer. This review aimed to identify whether people undergoing chemotherapy in different exercise interventions meet PA guidelines during chemotherapy. DATA SOURCES Databases searched were CINAHL complete, PubMed, Cochrane Library, EMBASE, AMED, Joanna Briggs Institute, OVID MEDLINE, and Google Scholar. Eligibility and risk-of-bias were reviewed by two authors. PROSPERO registration: CRD42018093839. CONCLUSION Thirty-three studies were eligible (14 randomized controlled trials, six pre-post, and 13 cross-sectional studies), which included 2,722 people with cancer undergoing chemotherapy. Studies (n = 11/13) found PA interventions successfully increased or maintained PA participation levels, whereas PA declined without intervention. This review has identified that due to limited evidence it is not possible to determine the most appropriate intervention to improve PA for people undergoing chemotherapy. Despite the well-documented benefits of PA, most people undergoing chemotherapy do not achieve the PA guidelines. Clinicians should be aware that during treatment, participants may not meet PA guidelines even if they participate in an intervention. However, interventions may prevent PA levels from declining. Further research is required to determine the most effective approaches to increase PA levels while undergoing chemotherapy. IMPLICATIONS FOR NURSING PRACTICE Nurses are well placed to have conversations with patients undergoing chemotherapy regarding PA levels. PA should be considered as part of a treatment regimen. Tailored advice must be provided aiming to improve health outcomes. Referral to an exercise professional is best practice to improve the uptake of PA.
Collapse
Affiliation(s)
- Rebecca Cesnik
- PhD Candidate, ACT Health, ACT, Australia; Professor, Sport and Exercise Science, Faculty of Health, University of Canberra, Bruce, ACT, Australia; Assistant Professor, Prehabilitation, Activity, Cancer, Exercise and Survivorship (PACES) Research Group, University of Canberra, Bruce, ACT, Australia
| | - Kellie Toohey
- Professor, Sport and Exercise Science, Faculty of Health, University of Canberra, Bruce, ACT, Australia; Assistant Professor, Prehabilitation, Activity, Cancer, Exercise and Survivorship (PACES) Research Group, University of Canberra, Bruce, ACT, Australia.
| | - Nicole Freene
- Associate Professor, Physiotherapy, Faculty of Health, University of Canberra, Bruce, ACT, Australia; Associate Professor, Health Research Institute, University of Canberra, Bruce, ACT, Australia
| | - Brea Kunstler
- Research Fellow, BehaviourWorks, Monash University, Victoria, Australia
| | - Stuart Semple
- Professor, Sport and Exercise Science, Faculty of Health, University of Canberra, Bruce, ACT, Australia; Associate Professor, Health Research Institute, University of Canberra, Bruce, ACT, Australia; Professor, Research Institute of Sport and Exercise, University of Canberra, Bruce, ACT, Australia
| |
Collapse
|
132
|
Bouabdallah S, Al-Maktoum A, Amin A. Steroidal Saponins: Naturally Occurring Compounds as Inhibitors of the Hallmarks of Cancer. Cancers (Basel) 2023; 15:3900. [PMID: 37568716 PMCID: PMC10417465 DOI: 10.3390/cancers15153900] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is a global health burden responsible for an exponentially growing number of incidences and mortalities, regardless of the significant advances in its treatment. The identification of the hallmarks of cancer is a major milestone in understanding the mechanisms that drive cancer initiation, development, and progression. In the past, the hallmarks of cancer have been targeted to effectively treat various types of cancers. These conventional cancer drugs have shown significant therapeutic efficacy but continue to impose unfavorable side effects on patients. Naturally derived compounds are being tested in the search for alternative anti-cancer drugs. Steroidal saponins are a group of naturally occurring compounds that primarily exist as secondary metabolites in plant species. Recent studies have suggested that steroidal saponins possess significant anti-cancer capabilities. This review aims to summarize the recent findings on steroidal saponins as inhibitors of the hallmarks of cancer and covers key studies published between the years 2014 and 2024. It is reported that steroidal saponins effectively inhibit the hallmarks of cancer, but poor bioavailability and insufficient preclinical studies limit their utilization.
Collapse
Affiliation(s)
- Salwa Bouabdallah
- Theranostic Biomarkers, LR23ES02, Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis 1006, Tunisia
| | - Amna Al-Maktoum
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Amr Amin
- Biology Department, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| |
Collapse
|
133
|
Aguilar R, Khan L, Arslanovic N, Birmingham K, Kasliwal K, Posnikoff S, Chakraborty U, Hickman AR, Watson R, Ezell RJ, Willis HE, Cowles MW, Garner R, Shim A, Gutierrez I, Marunde MR, Keogh MC, Tyler JK. Multivalent binding of the tardigrade Dsup protein to chromatin promotes yeast survival and longevity upon exposure to oxidative damage. RESEARCH SQUARE 2023:rs.3.rs-3182883. [PMID: 37546815 PMCID: PMC10402244 DOI: 10.21203/rs.3.rs-3182883/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Tardigrades are remarkable in their ability to survive extreme environments. The damage suppressor (Dsup) protein is thought responsible for their extreme resistance to reactive oxygen species (ROS) generated by irradiation. Here we show that expression of Ramazzottius varieornatus Dsup in Saccharomyces cerevisiae reduces oxidative DNA damage and extends the lifespan of budding yeast exposed to chronic oxidative genotoxicity. This protection from ROS requires either the Dsup HMGN-like domain or sequences C-terminal to same. Dsup associates with no apparent bias across the yeast genome, using multiple modes of nucleosome binding; the HMGN-like region interacts with both the H2A/H2B acidic patch and H3/H4 histone tails, while the C-terminal region binds DNA. These findings give precedent for engineering an organism by physically shielding its genome to promote survival and longevity in the face of oxidative damage.
Collapse
Affiliation(s)
- Rhiannon Aguilar
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | | | - Nina Arslanovic
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
| | - Kaylah Birmingham
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
- Weill Cornell Medicine, Pharmacology Graduate Program, New York, NY 10065 United States
| | - Kritika Kasliwal
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
- Weill Cornell Medicine, Biochemistry, Cellular, and Molecular Biology Graduate Program, New York, NY 10065, USA
| | - Spike Posnikoff
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
| | - Ujani Chakraborty
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
| | | | | | | | | | | | - Richard Garner
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
- Weill Cornell Medicine, Biochemistry, Cellular, and Molecular Biology Graduate Program, New York, NY 10065, USA
| | - Abraham Shim
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
- Weill Cornell Medicine, Biochemistry, Cellular, and Molecular Biology Graduate Program, New York, NY 10065, USA
| | - Ignacio Gutierrez
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
| | | | | | - Jessica K. Tyler
- Weill Cornell Medicine, Department of Pathology and Laboratory Medicine, New York, NY 10065, USA
| |
Collapse
|
134
|
Kisamore CO, Elliott BD, DeVries AC, Nelson RJ, Walker WH. Chronotherapeutics for Solid Tumors. Pharmaceutics 2023; 15:2023. [PMID: 37631237 PMCID: PMC10459260 DOI: 10.3390/pharmaceutics15082023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Circadian rhythms are internal manifestations of the 24-h solar day that allow for synchronization of biological and behavioral processes to the external solar day. This precise regulation of physiology and behavior improves adaptive function and survival. Chronotherapy takes advantage of circadian rhythms in physiological processes to optimize the timing of drug administration to achieve maximal therapeutic efficacy and minimize negative side effects. Chronotherapy for cancer treatment was first demonstrated to be beneficial more than five decades ago and has favorable effects across diverse cancer types. However, implementation of chronotherapy in clinic remains limited. The present review examines the evidence for chronotherapeutic treatment for solid tumors. Specifically, studies examining chrono-chemotherapy, chrono-radiotherapy, and alternative chronotherapeutics (e.g., hormone therapy, TKIs, antiangiogenic therapy, immunotherapy) are discussed. In addition, we propose areas of needed research and identify challenges in the field that remain to be addressed.
Collapse
Affiliation(s)
- Claire O. Kisamore
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (C.O.K.); (B.D.E.); (A.C.D.); (R.J.N.)
| | - Brittany D. Elliott
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (C.O.K.); (B.D.E.); (A.C.D.); (R.J.N.)
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (C.O.K.); (B.D.E.); (A.C.D.); (R.J.N.)
- Department of Medicine, West Virginia University, Morgantown, WV 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (C.O.K.); (B.D.E.); (A.C.D.); (R.J.N.)
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| | - William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA; (C.O.K.); (B.D.E.); (A.C.D.); (R.J.N.)
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
| |
Collapse
|
135
|
Jeewon R, Aullybux AA, Puchooa D, Nazurally N, Alrefaei AF, Zhang Y. Marine Microbial Polysaccharides: An Untapped Resource for Biotechnological Applications. Mar Drugs 2023; 21:420. [PMID: 37504951 PMCID: PMC10381399 DOI: 10.3390/md21070420] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
As the largest habitat on Earth, the marine environment harbors various microorganisms of biotechnological potential. Indeed, microbial compounds, especially polysaccharides from marine species, have been attracting much attention for their applications within the medical, pharmaceutical, food, and other industries, with such interest largely stemming from the extensive structural and functional diversity displayed by these natural polymers. At the same time, the extreme conditions within the aquatic ecosystem (e.g., temperature, pH, salinity) may not only induce microorganisms to develop a unique metabolism but may also increase the likelihood of isolating novel polysaccharides with previously unreported characteristics. However, despite their potential, only a few microbial polysaccharides have actually reached the market, with even fewer being of marine origin. Through a synthesis of relevant literature, this review seeks to provide an overview of marine microbial polysaccharides, including their unique characteristics. In particular, their suitability for specific biotechnological applications and recent progress made will be highlighted before discussing the challenges that currently limit their study as well as their potential for wider applications. It is expected that this review will help to guide future research in the field of microbial polysaccharides, especially those of marine origin.
Collapse
Affiliation(s)
- Rajesh Jeewon
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Aadil Ahmad Aullybux
- Department of Agricultural and Food Science, Faculty of Agriculture, University of Mauritius, Réduit 80837, Mauritius
| | - Daneshwar Puchooa
- Department of Agricultural and Food Science, Faculty of Agriculture, University of Mauritius, Réduit 80837, Mauritius
| | - Nadeem Nazurally
- Department of Agricultural and Food Science, Faculty of Agriculture, University of Mauritius, Réduit 80837, Mauritius
| | - Abdulwahed Fahad Alrefaei
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ying Zhang
- School of Ecology and Natural Conservation, Beijing Forestry University, 35 East Qinghua Road, Haidian District, Beijing 100083, China
| |
Collapse
|
136
|
Seo H, Verma A, Kinzel M, Huang Q, Mahoney DJ, Jacquelot N. Targeting Potential of Innate Lymphoid Cells in Melanoma and Other Cancers. Pharmaceutics 2023; 15:2001. [PMID: 37514187 PMCID: PMC10384206 DOI: 10.3390/pharmaceutics15072001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Reinvigorating the killing function of tumor-infiltrating immune cells through the targeting of regulatory molecules expressed on lymphocytes has markedly improved the prognosis of cancer patients, particularly in melanoma. While initially thought to solely strengthen adaptive T lymphocyte anti-tumor activity, recent investigations suggest that other immune cell subsets, particularly tissue-resident innate lymphoid cells (ILCs), may benefit from immunotherapy treatment. Here, we describe the recent findings showing immune checkpoint expression on tissue-resident and tumor-infiltrating ILCs and how their effector function is modulated by checkpoint blockade-based therapies in cancer. We discuss the therapeutic potential of ILCs beyond the classical PD-1 and CTLA-4 regulatory molecules, exploring other possibilities to manipulate ILC effector function to further impede tumor growth and quench disease progression.
Collapse
Affiliation(s)
- Hobin Seo
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Amisha Verma
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Kinzel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Qiutong Huang
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, QLD 4102, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Nicolas Jacquelot
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
137
|
El-Tanani M, Nsairat H, Matalka II, Aljabali AAA, Mishra V, Mishra Y, Naikoo GA, Chava SR, Charbe NB, Tambuwala MM. Impact of exosome therapy on pancreatic cancer and its progression. Med Oncol 2023; 40:225. [PMID: 37405480 PMCID: PMC10322774 DOI: 10.1007/s12032-023-02101-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/19/2023] [Indexed: 07/06/2023]
Abstract
Pancreatic cancer, one of the most aggressive tumors, has a dismal prognosis because of the low rates of early identification, fast progression, difficulties following surgery, and the ineffectiveness of current oncologic therapies. There are no imaging techniques or biomarkers that can accurately identify, categorize, or predict the biological behavior of this tumor. Exosomes are extracellular vesicles that play a crucial rule in the progression, metastasis, and chemoresistance of pancreatic cancer. They have been verified to be potential biomarkers for pancreatic cancer management. Studying the role of exosomes in pancreatic cancer is substantial. Exosomes are secreted by most eukaryotic cells and participated in intercellular communication. The components of exosomes, including proteins, DNA, mRNA, microRNA, long non-coding RNA, circular RNA, etc., play a crucial role in regulating tumor growth, metastasis, and angiogenesis in the process of cancer development, and can be used as a prognostic marker and/or grading basis for tumor patients. Hereby, in this concise review, we intend to summarize exosomes components and isolation, exosome secretion, function, importance of exosomes in the progression of pancreatic cancer and exosomal miRNAs as possible pancreatic cancer biomarkers. Finally, the application potential of exosomes in the treatment of pancreatic cancer, which provides theoretical supports for using exosomes to serve precise tumor treatment in the clinic, will be discussed.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, West Yorkshire, UK.
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid, 21163, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, PC 211, Salalah, Oman
| | | | - Nitin B Charbe
- Department of Pharmaceutics, College of Pharmacy, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|
138
|
Garcés-Garcés J, Sánchez-Martos M, Martinez-Navarrete G, Fernández-Jover E, Encheva M, León M, Ortiz J, Sastre-Santos Á, Fernández-Lázaro F. New Highly Fluorescent Water Soluble Imidazolium-Perylenediimides: Synthesis and Cellular Response. Pharmaceutics 2023; 15:1892. [PMID: 37514077 PMCID: PMC10384807 DOI: 10.3390/pharmaceutics15071892] [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: 05/17/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The synthesis and characterization of two new water soluble 2,6-bis(imidazolylmethyl)-4-methylphenoxy-containing perylenediimides, PDI-1 and PDI-2, are described. These compounds demonstrate a high fluorescence quantum yield in water and were investigated as potential photosensitizers for generating reactive oxygen species with applications in anticancer activities. The HeLa cell line (VPH18) was used to evaluate their efficacy. Fluorescence microscopy was employed to confirm the successful internalization of PDI-1 and PDI-2, while confocal microscopy revealed the specific locations of both PDIs within the lysosomes and mitochondria. In vitro studies were conducted to evaluate the anticancer activity of PDI-1 and PDI-2. Remarkably, these photosensitizers demonstrated a significant ability to selectively eliminate cancer cells when exposed to a specific light wavelength. The water solubility, high fluorescence quantum yield, and selective cytotoxicity of these PDIs toward cancer cells highlight their potential as effective agents for targeted photodynamic therapy. In conclusion, the findings presented here provide a strong foundation for the future exploration and optimization of PDI-1 and PDI-2 as effective photosensitizers in photodynamic therapy, potentially leading to improved treatment strategies for cancer patients.
Collapse
Affiliation(s)
- José Garcés-Garcés
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Miguel Sánchez-Martos
- Área de Neuroprótesis y Rehabilitación Visual, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Gema Martinez-Navarrete
- Área de Neuroprótesis y Rehabilitación Visual, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Eduardo Fernández-Jover
- Área de Neuroprótesis y Rehabilitación Visual, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Mirela Encheva
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Martín León
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Javier Ortiz
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Ángela Sastre-Santos
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| | - Fernando Fernández-Lázaro
- Área de Química Orgánica, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avda. de la Universidad s/n, 03202 Elche, Spain
| |
Collapse
|
139
|
Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023; 11:1879. [PMID: 37509518 PMCID: PMC10377337 DOI: 10.3390/biomedicines11071879] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inhibition of the interaction between MDM2 and p53 has emerged as a promising strategy for combating cancer, including the treatment of glioblastoma (GBM). Numerous MDM2 inhibitors have been developed and are currently undergoing rigorous testing for their potential in GBM therapy. Encouraging results from studies conducted in cell culture and animal models suggest that MDM2 inhibitors could effectively treat a specific subset of GBM patients with wild-type TP53 or functional p53. Combination therapy with clinically established treatment modalities such as radiation and chemotherapy offers the potential to achieve a more profound therapeutic response. Furthermore, an increasing array of other molecularly targeted therapies are being explored in combination with MDM2 inhibitors to increase the effects of individual treatments. While some MDM2 inhibitors have progressed to early phase clinical trials in GBM, their efficacy, alone and in combination, is yet to be confirmed. In this article, we present an overview of MDM2 inhibitors currently under preclinical and clinical investigation, with a specific focus on the drugs being assessed in ongoing clinical trials for GBM patients.
Collapse
Affiliation(s)
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Leo F Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
140
|
AbdulHussein AH, Al-Taee MM, Radih ZA, Aljuboory DS, Mohammed ZQ, Hashesh TS, Riadi Y, Hadrawi SK, Najafi M. Mechanisms of cancer cell death induction by triptolide. Biofactors 2023; 49:718-735. [PMID: 36876465 DOI: 10.1002/biof.1944] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/07/2023]
Abstract
Drug resistance is a hot topic issue in cancer research and therapy. Although cancer therapy including radiotherapy and anti-cancer drugs can kill malignant cells within the tumor, cancer cells can develop a wide range of mechanisms to resist the toxic effects of anti-cancer agents. Cancer cells may provide some mechanisms to resist oxidative stress and escape from apoptosis and attack by the immune system. Furthermore, cancer cells may resist senescence, pyroptosis, ferroptosis, necroptosis, and autophagic cell death by modulating several critical genes. The development of these mechanisms leads to resistance to anti-cancer drugs and also radiotherapy. Resistance to therapy can increase mortality and reduce survival following cancer therapy. Thus, overcoming mechanisms of resistance to cell death in malignant cells can facilitate tumor elimination and increase the efficiency of anti-cancer therapy. Natural-derived molecules are intriguing agents that may be suggested to be used as an adjuvant in combination with other anticancer drugs or radiotherapy to sensitize cancer cells to therapy with at least side effects. This paper aims to review the potential of triptolide for inducing various types of cell death in cancer cells. We review the induction or resistance to different cell death mechanisms such as apoptosis, autophagic cell death, senescence, pyroptosis, ferroptosis, and necrosis following the administration of triptolide. We also review the safety and future perspectives for triptolide and its derivatives in experimental and human studies. The anticancer potential of triptolide and its derivatives may make them effective adjuvants for enhancing tumor suppression in combination with anticancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Salema K Hadrawi
- Refrigeration and Air-Conditioning Technical Engineering Department, College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
141
|
Farhan M. Insights on the Role of Polyphenols in Combating Cancer Drug Resistance. Biomedicines 2023; 11:1709. [PMID: 37371804 PMCID: PMC10296548 DOI: 10.3390/biomedicines11061709] [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: 05/22/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chemotherapy resistance is still a serious problem in the treatment of most cancers. Many cellular and molecular mechanisms contribute to both inherent and acquired drug resistance. They include the use of unaffected growth-signaling pathways, changes in the tumor microenvironment, and the active transport of medicines out of the cell. The antioxidant capacity of polyphenols and their potential to inhibit the activation of procarcinogens, cancer cell proliferation, metastasis, and angiogenesis, as well as to promote the inhibition or downregulation of active drug efflux transporters, have been linked to a reduced risk of cancer in epidemiological studies. Polyphenols also have the ability to alter immunological responses and inflammatory cascades, as well as trigger apoptosis in cancer cells. The discovery of the relationship between abnormal growth signaling and metabolic dysfunction in cancer cells highlights the importance of further investigating the effects of dietary polyphenols, including their ability to boost the efficacy of chemotherapy and avoid multidrug resistance (MDR). Here, it is summarized what is known regarding the effectiveness of natural polyphenolic compounds in counteracting the resistance that might develop to cancer drugs as a result of a variety of different mechanisms.
Collapse
Affiliation(s)
- Mohd Farhan
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Ahsa 31982, Saudi Arabia
| |
Collapse
|
142
|
Gariboldi MB, Marras E, Ferrario N, Vivona V, Prini P, Vignati F, Perletti G. Anti-Cancer Potential of Edible/Medicinal Mushrooms in Breast Cancer. Int J Mol Sci 2023; 24:10120. [PMID: 37373268 DOI: 10.3390/ijms241210120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Edible/medicinal mushrooms have been traditionally used in Asian countries either in the cuisine or as dietary supplements and nutraceuticals. In recent decades, they have aroused increasing attention in Europe as well, due to their health and nutritional benefits. In particular, among the different pharmacological activities reported (antibacterial, anti-inflammatory, antioxidative, antiviral, immunomodulating, antidiabetic, etc.), edible/medicinal mushrooms have been shown to exert in vitro and in vivo anticancer effects on several kinds of tumors, including breast cancer. In this article, we reviewed mushrooms showing antineoplastic activity again breast cancer cells, especially focusing on the possible bioactive compounds involved and their mechanisms of action. In particular, the following mushrooms have been considered: Agaricus bisporus, Antrodia cinnamomea, Cordyceps sinensis, Cordyceps militaris, Coriolus versicolor, Ganoderma lucidum, Grifola frondosa, Lentinula edodes, and Pleurotus ostreatus. We also report insights into the relationship between dietary consumption of edible mushrooms and breast cancer risk, and the results of clinical studies and meta-analyses focusing on the effects of fungal extracts on breast cancer patients.
Collapse
Affiliation(s)
- Marzia Bruna Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Emanuela Marras
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Nicole Ferrario
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Pamela Prini
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Francesca Vignati
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| | - Gianpaolo Perletti
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, 21100 Varese, Italy
| |
Collapse
|
143
|
Mitxelena J, Zubiaga AM. Foreword Special Issue Genomic Instability in Tumor Evolution and Therapy Response. Cancers (Basel) 2023; 15:3080. [PMID: 37370691 DOI: 10.3390/cancers15123080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
From an evolutionary perspective, mutations in the DNA molecule act as a source of genetic variation and thus, are beneficial to the adaptation and survival of the species [...].
Collapse
Affiliation(s)
- Jone Mitxelena
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ana M Zubiaga
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain
| |
Collapse
|
144
|
Du K, Wang L, Wang Z, Xiao H, Hou J, Hu L, Fan N, Wang Y. Angelica Sinensis polysaccharide antagonizes 5-Fluorouracil-induced spleen injury and dysfunction by suppressing oxidative stress and apoptosis. Biomed Pharmacother 2023; 162:114602. [PMID: 37018993 DOI: 10.1016/j.biopha.2023.114602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Angelica Sinensis polysaccharide (ASP), the main active component of Angelica sinensis, possesses antioxidative and anti-apoptotic properties. In this study, we have investigated the antagonistic effect of ASP on 5-FU-induced injury of mouse spleen in vivo and splenocytes in vitro, and its possible mechanism. Our results showed that ASP inhibited 5-FU-induced decreases in spleen weight and organ index in mice, restored the number of peripheral blood leukocytes and lymphocytes, repaired spleen structure disorder and functional impairment, rescued serum IL-2, IL-6, and IFN-γ levels, and relieved 5-FU-induced mitochondrial swelling, reduced the oxidant accumulation including MDA and ROS, whereas increasing the activities of GSH, SOD and CAT. The mechanism may be related to ASP downregulation of Keap1 protein expression thus motivating the nuclear translocation of Nrf2. Furthermore, ASP alleviated the apoptosis of spleens in vivo and splenocytes in vitro, and reactivated PI3K / AKT signalling. In conclusion, the protective effect of ASP on spleens and splenocytes may be related to the reduction of oxidative stress and apoptosis via reactivation of Nrf2 and PI3K/AKT pathways. This study has provided a new protective agent for minimizing the spleen injury caused by 5-FU and a new idea for improving the prognosis of chemotherapy patients.
Collapse
Affiliation(s)
- Kunhang Du
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Lu Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ziling Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Hanxianzhi Xiao
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Jiying Hou
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ling Hu
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ningke Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
145
|
Menéndez-Velázquez A, García-Delgado AB. A Novel Photopharmacological Tool: Dual-Step Luminescence for Biological Tissue Penetration of Light and the Selective Activation of Photodrugs. Int J Mol Sci 2023; 24:ijms24119404. [PMID: 37298355 DOI: 10.3390/ijms24119404] [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: 04/15/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Conventional pharmacology lacks spatial and temporal selectivity in terms of drug action. This leads to unwanted side effects, such as damage to healthy cells, as well as other less obvious effects, such as environmental toxicity and the acquisition of resistance to drugs, especially antibiotics, by pathogenic microorganisms. Photopharmacology, based on the selective activation of drugs by light, can contribute to alleviating this serious problem. However, many of these photodrugs are activated by light in the UV-visible spectral range, which does not propagate through biological tissues. In this article, to overcome this problem, we propose a dual-spectral conversion technique, which simultaneously makes use of up-conversion (using rare earth elements) and down-shifting (using organic materials) techniques in order to modify the spectrum of light. Near-infrared light (980 nm), which penetrates tissue fairly well, can provide a "remote control" for drug activation. Once near-IR light is inside the body, it is up-converted to the UV-visible spectral range. Subsequently, this radiation is down-shifted in order to accurately adjust to the excitation wavelengths of light which can selectively activate hypothetical and specific photodrugs. In summary, this article presents, for the first time, a "dual tunable light source" which can penetrate into the human body and deliver light of specific wavelengths; thus, it can overcome one of the main limitations of photopharmacology. It opens up promising possibilities for the moving of photodrugs from the laboratory to the clinic.
Collapse
|
146
|
Li X, Huang Z, Liao Z, Liu A, Huo S. Transformable nanodrugs for overcoming the biological barriers in the tumor environment during drug delivery. NANOSCALE 2023; 15:8532-8547. [PMID: 37114478 DOI: 10.1039/d2nr06621a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Drug delivery systems have been studied massively with explosive growth in the last few decades. However, challenges such as biological barriers are still obstructing the delivery efficiency of nanomedicines. Reports have shown that the physicochemical properties, such as the morphologies of nanodrugs, could highly affect their biodistribution and bioavailability. Therefore, transformable nanodrugs that take advantage of different sizes and shapes allow for overcoming multiple biological barriers, providing promising prospects for drug delivery. This review aims to present an overview of the most recent developments of transformable nanodrugs in this emerging field. First, the design principles and transformation mechanisms which serve as guidelines for smart nanodrugs are summarized. Afterward, their applications in overcoming biological barriers, including the bloodstream, intratumoral pressure, cellular membrane, endosomal wrapping, and nuclear membrane, are highlighted. Finally, discussions on the current developments and future perspectives of transformable nanodrugs are given.
Collapse
Affiliation(s)
- Xuejian Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhenkun Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Aijie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
147
|
Michalak O, Cybulski M, Szymanowski W, Gornowicz A, Kubiszewski M, Ostrowska K, Krzeczyński P, Bielawski K, Trzaskowski B, Bielawska A. Synthesis, Biological Activity, ADME and Molecular Docking Studies of Novel Ursolic Acid Derivatives as Potent Anticancer Agents. Int J Mol Sci 2023; 24:ijms24108875. [PMID: 37240221 DOI: 10.3390/ijms24108875] [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: 03/30/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
A series of new ursolic acid (UA) derivatives substituted with various amino acids (AAs) or dipeptides (DP) at the C-3 position of the steroid skeleton was designed and synthesized. The compounds were obtained by the esterification of UA with the corresponding AAs. The cytotoxic activity of the synthesized conjugates was determined using the hormone-dependent breast cancer cell line MCF-7 and the triple-negative breast cancer cell line MDA. Three derivatives (l-seryloxy-, l-prolyloxy- and l-alanyl-l-isoleucyloxy-) showed micromolar IC50 values and reduced the concentrations of matrix metalloproteinases 2 and 9. Further studies revealed that for two compounds (l-seryloxy- and l-alanyl-l-isoleucyloxy-), a possible mechanism of their antiproliferative action is the activation of caspase-7 and the proapoptotic Bax protein in the apoptotic pathway. The third compound (l-prolyloxy- derivative) showed a different mechanism of action as it induced autophagy as measured by an increase in the concentrations of three autophagy markers: LC3A, LC3B, and beclin-1. This derivative also showed statistically significant inhibition of the proinflammatory cytokines TNF-α and IL-6. Finally, for all synthesized compounds, we computationally predicted their ADME properties as well as performed molecular docking to the estrogen receptor to assess their potential for further development as anticancer agents.
Collapse
Affiliation(s)
- Olga Michalak
- Chemistry Section, Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - Marcin Cybulski
- Chemistry Section, Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - Wojciech Szymanowski
- Department of Biotechnology, Medical University of Bialystok, 1 Kilińskiego Str., 15-089 Bialystok, Poland
| | - Agnieszka Gornowicz
- Department of Biotechnology, Medical University of Bialystok, 1 Kilińskiego Str., 15-089 Bialystok, Poland
| | - Marek Kubiszewski
- Analytical Research Section, Pharmaceutical Analysis Laboratory, Łukasiewicz Research Network-Industrial Chemistry Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - Kinga Ostrowska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Piotr Krzeczyński
- Chemistry Section, Pharmacy, Cosmetic Chemistry and Biotechnology Research Group, Łukasiewicz Research Network-Industrial Chemistry Institute, 8 Rydygiera Str., 01-793 Warsaw, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, 1 Kilińskiego Str., 15-089 Bialystok, Poland
| | - Bartosz Trzaskowski
- Chemical and Biological Systems Simulation Lab, Center of New Technologies, University of Warsaw, 2C Banacha Str., 02-097 Warsaw, Poland
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, 1 Kilińskiego Str., 15-089 Bialystok, Poland
| |
Collapse
|
148
|
Chen C, Hu M, Cao Y, Zhu B, Chen J, Li Y, Shao J, Zhou S, Shan P, Zheng C, Li Z, Li Z. Combination of a STING Agonist and Photothermal Therapy Using Chitosan Hydrogels for Cancer Immunotherapy. Biomacromolecules 2023. [PMID: 37125731 DOI: 10.1021/acs.biomac.3c00196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cyclic dinucleotides (CDNs) are a promising class of immune agonists that trigger the stimulator of interferon genes (STING) to activate both innate and acquired immunity. However, the efficacy of CDNs is limited by drug delivery barriers. Therefore, we developed a combined immunotherapy strategy based on injectable reactive oxygen species (ROS)-responsive hydrogels, which sustainably release 5,6-dimethylxanthenone-4-acetic acid (DMXAA) as known as a STING agonist and indocyanine green (ICG) by utilizing a high level of ROS in the tumor microenvironment (TME). The STING agonist combined with photothermal therapy (PTT) can improve the biological efficacy of DMXAA, transform the immunosuppressive TME into an immunogenic and tumoricidal microenvironment, and completely kill tumor cells. In addition, this bioreactive gel can effectively leverage local ROS to facilitate the release of immunotherapy drugs, thereby enhancing the efficacy of combination therapy, improving the TME, inhibiting tumor growth, inducing memory immunity, and protecting against tumor rechallenge.
Collapse
Affiliation(s)
- Cunguo Chen
- Department of Dermatology and Venereology, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang 325200, P. R. China
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Murong Hu
- Department of Dermatology and Venereology, Hangzhou Third Hospital, Hangzhou, Zhejiang 321000, P. R. China
| | - Yunyun Cao
- Nursing Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Binbin Zhu
- Nursing Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Jiashe Chen
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Yashi Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Junyi Shao
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Sen Zhou
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Pengfei Shan
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou 325027, P. R. China
| | - Chen Zheng
- Department of Breast Cancer Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Zhongyu Li
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou 325027, P. R. China
| | - Zhiming Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| |
Collapse
|
149
|
Zuhrotun A, Oktaviani DJ, Hasanah AN. Biosynthesis of Gold and Silver Nanoparticles Using Phytochemical Compounds. Molecules 2023; 28:molecules28073240. [PMID: 37050004 PMCID: PMC10096681 DOI: 10.3390/molecules28073240] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023] Open
Abstract
Gold and silver nanoparticles are nanoparticles that have been widely used in various fields and have shown good benefits. The method of nanoparticle biosynthesis utilizing plant extracts, also known as green synthesis, has become a promising method considering the advantages it has compared to other synthesis methods. This review aims to give an overview of the phytochemical compounds in plants used in the synthesis of gold and silver nanoparticles, the nanoparticle properties produced using plant extracts based on the concentration and structure of phytochemical compounds, and their applications. Phytochemical compounds play an important role as reducing agents and stabilizers in the stages of the synthesis of nanoparticles. Polyphenol compounds, reducing sugars, and proteins are the main phytochemical compounds that are responsible for the synthesis of gold and silver nanoparticles. The concentration of phytochemical compounds affects the physical properties, stability, and activity of nanoparticles. This is important to know to be able to overcome limitations in controlling the physical properties of the nanoparticles produced. Based on structure, the phytochemical compounds that have ortho-substituted hydroxyl result in a smaller size and well-defined shape, which can lead to greater activity and stability. Furthermore, the optimal condition of the biosynthesis process is required to gain a successful reaction that includes setting the metal ion concentration, temperature, reaction time, and pH.
Collapse
Affiliation(s)
- Ade Zuhrotun
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Jalan Raya Bandung-Sumedang KM 21 Jatinangor, Bandung 45363, Indonesia
| | - Dede Jihan Oktaviani
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Jalan Raya Bandung-Sumedang KM 21 Jatinangor, Bandung 45363, Indonesia
| | - Aliya Nur Hasanah
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jalan Raya Bandung-Sumedang KM 21 Jatinangor, Bandung 45363, Indonesia
| |
Collapse
|
150
|
Roldán-Peña JM, Puerta A, Dinić J, Jovanović Stojanov S, González-Bakker A, Hicke FJ, Mishra A, Piyasaengthong A, Maya I, Walton JW, Pešić M, Padrón JM, Fernández-Bolaños JG, López Ó. Biotinylated selenocyanates: Potent and selective cytostatic agents. Bioorg Chem 2023; 133:106410. [PMID: 36822000 DOI: 10.1016/j.bioorg.2023.106410] [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: 12/08/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Most of the currently available cytotoxic agents for tackling cancer are devoid of selectivity, thus causing severe side-effects. This situation stimulated us to develop new antiproliferative agents with enhanced affinity towards tumour cells. We focused our attention on novel chalcogen-containing compounds (thiosemicarbazones, disulfides, selenoureas, thio- and selenocyanates), and particularly on selenium derivatives, as it has been documented that this kind of compounds might act as prodrugs releasing selenium-based reactive species on tumour cells. Particularly interesting in terms of potency and selectivity was a pharmacophore comprised by a selenocyanato-alkyl fragment connected to a p-phenylenediamine residue, where the nature of the second amino moiety (free, Boc-protected, enamine-protected) provided a wide variety of antiproliferative activities, ranging from the low micromolar to the nanomolar values. The optimized structure was in turn conjugated through a peptide linkage with biotin (vitamin B7), a cellular growth promoter, whose receptor is overexpressed in numerous cancer cells; the purpose was to develop a selective vector towards malignant cells. Such biotinylated derivative behaved as a very strong antiproliferative agent, achieving GI50 values in the low nM range for most of the tested cancer cells; moreover, it was featured with an outstanding selectivity, with GI50 > 100 µM against human fibroblasts. Mechanistic studies on the mode of inhibition of the biotinylated selenocyanate revealed (Annexin-V assay) a remarkable increase in the number of apoptotic cells compared to the control experiment; moreover, depolarization of the mitochondrial membrane was detected by flow cytometry analysis, and with fluorescent microscopy, what supports the apoptotic cell death. Prior to the apoptotic events, cytostatic effects were observed against SW1573 cells using label-free cell-living imaging; therefore, tumour cell division was prevented. Multidrug resistant cell lines exhibited a reduced sensitivity towards the biotinylated selenocyanate, probably due to its P-gp-mediated efflux. Remarkably, antiproliferative levels could be restored by co-administration with tariquidar, a P-gp inhibitor; this approach can, therefore, overcome multidrug resistance mediated by the P-gp efflux system.
Collapse
Affiliation(s)
- Jesús M Roldán-Peña
- Organic Chemistry Department, Faculty of Chemistry, University of Seville, PO box 1203, E-41071 Seville, Spain
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain
| | - Jelena Dinić
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Sofija Jovanović Stojanov
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia
| | - Aday González-Bakker
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain
| | - Francisco J Hicke
- Organic Chemistry Department, Faculty of Chemistry, University of Seville, PO box 1203, E-41071 Seville, Spain
| | - Atreyee Mishra
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK
| | - Akkharadet Piyasaengthong
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK; Bioscience Program, Faculty of Science, Kasetsart University, Bangkok 10900, Chatuchak, Thailand
| | - Inés Maya
- Organic Chemistry Department, Faculty of Chemistry, University of Seville, PO box 1203, E-41071 Seville, Spain
| | - James W Walton
- Department of Chemistry, Durham University, Lower Mountjoy, South Road, Durham DH1 3LE, UK
| | - Milica Pešić
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060 Belgrade, Serbia.
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, Astrofísico Francisco Sánchez 2, E-38206 La Laguna, Spain.
| | - José G Fernández-Bolaños
- Organic Chemistry Department, Faculty of Chemistry, University of Seville, PO box 1203, E-41071 Seville, Spain.
| | - Óscar López
- Organic Chemistry Department, Faculty of Chemistry, University of Seville, PO box 1203, E-41071 Seville, Spain.
| |
Collapse
|