1
|
Karankar VS, Awasthi S, Srivastava N. Peptide-driven strategies against lung cancer. Life Sci 2025; 366-367:123453. [PMID: 39923837 DOI: 10.1016/j.lfs.2025.123453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Lung cancer remains one of the most significant global health challenges, accounting for 18 % of all cancer-related deaths. While risk factors such as heavy metal exposure and cigarette smoking are well-known contributors, the limitations of conventional treatments including severe side effects and drug resistance highlight the urgent need for more targeted and safer therapeutic options. In this context, peptides have emerged as a novel, precise, and effective class of therapies for lung cancer treatment. They have shown promise in limiting lung cancer progression by targeting key molecular pathways involved in tumour growth. Anti-non-small cell lung cancer peptides that specifically target proteins such as EGFR, TP53, BRAF, MET, ROS1, and ALK have demonstrated potential in improving lung cancer outcomes. Additionally, anti-inflammatory and apoptosis-inducing peptides offer further therapeutic benefits. This review provides a comprehensive overview of the peptides currently in use or under investigation for the treatment of lung cancer, highlighting their mechanisms of action and therapeutic potential. As research continues to advance, peptides are poised to become a promising new therapeutic option in the fight against lung cancer.
Collapse
Affiliation(s)
- Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
2
|
Garaci E, Paci M, Matteucci C, Costantini C, Puccetti P, Romani L. Phenotypic drug discovery: a case for thymosin alpha-1. Front Med (Lausanne) 2024; 11:1388959. [PMID: 38903817 PMCID: PMC11187271 DOI: 10.3389/fmed.2024.1388959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
Phenotypic drug discovery (PDD) involves screening compounds for their effects on cells, tissues, or whole organisms without necessarily understanding the underlying molecular targets. PDD differs from target-based strategies as it does not require knowledge of a specific drug target or its role in the disease. This approach can lead to the discovery of drugs with unexpected therapeutic effects or applications and allows for the identification of drugs based on their functional effects, rather than through a predefined target-based approach. Ultimately, disease definitions are mostly symptom-based rather than mechanism-based, and the therapeutics should be likewise. In recent years, there has been a renewed interest in PDD due to its potential to address the complexity of human diseases, including the holistic picture of multiple metabolites engaging with multiple targets constituting the central hub of the metabolic host-microbe interactions. Although PDD presents challenges such as hit validation and target deconvolution, significant achievements have been reached in the era of big data. This article explores the experiences of researchers testing the effect of a thymic peptide hormone, thymosin alpha-1, in preclinical and clinical settings and discuss how its therapeutic utility in the precision medicine era can be accommodated within the PDD framework.
Collapse
Affiliation(s)
| | - Maurizio Paci
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- San Raffaele Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Vaezi-Kakhki A, Asoodeh A. Comparison of different methods for synthesis of iron oxide nanoparticles and investigation of their cellular properties, and antioxidant potential. Int J Pharm 2023; 645:123417. [PMID: 37714316 DOI: 10.1016/j.ijpharm.2023.123417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/03/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Iron oxide nanoparticles could play a useful role in lung cancer therapy. Iron oxide nanoparticles (NPs) were synthesized by plant mediated synthesis, chemical, and microbial mediated synthesis. iron oxide nanoparticle polyethylene glycol cis-diamminedichloroplatinum (Fe2O3@PEG@CDDP(, iron oxide nanoparticle polyethylene glycol (Fe2O3@PEG), and cis-diamminedichloroplatinum (CDDP) were evaluated for their antioxidant,and in vitro cytotoxicity tests. X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), Field emission scanning electron microscopy (FE-SEM), mapping, and zeta potential were used to characterize the synthesized iron oxides NPs. Cell toxicity was determined using A549 and HFF cells by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. The antioxidant scavenging activity of Fe2O3@PEG@CDDP, Fe2O3@PEG, and CDDP displayed IC50 values (11.96, 26.74, and 3.17 μg/ml) and (8.54, 11.4, and 1.14 μg/ml) in 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2,2-Diphenyl-1-picrylhydrazyl (DPPH) assays, respectively. Nanoparticles obtained from plant mediated synthesis method showed the great antioxidant activity. Results showed that, green-method synthesized nanoparticles were the most effective at killing cancer cells. Thus, the characteristics of nanoparticles from green synthesis are more valuable than the other methods. Green synthesis is environmental friendly cost-effective, and easy approach for synthesize NPs.
Collapse
Affiliation(s)
- Abbas Vaezi-Kakhki
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
4
|
Liu Y, Lu J. Mechanism and clinical application of thymosin in the treatment of lung cancer. Front Immunol 2023; 14:1237978. [PMID: 37701432 PMCID: PMC10493777 DOI: 10.3389/fimmu.2023.1237978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The burden of cancer on public health is becoming more widely acknowledged. Lung cancer has one of the highest incidence and mortality rates of all cancers. The prevalence of early screening, the emergence of targeted therapy, and the development of immunotherapy have all significantly improved the overall prognosis of lung cancer patients. The current state of affairs, however, is not encouraging, and there are issues like poor treatment outcomes for some patients and extremely poor prognoses for those with advanced lung cancer. Because of their potent immunomodulatory capabilities, thymosin drugs are frequently used in the treatment of tumors. The effectiveness of thymosin drugs in the treatment of lung cancer has been demonstrated in numerous studies, which amply demonstrates the potential and future of thymosin drugs for the treatment of lung cancer. The clinical research on thymosin peptide drugs in lung cancer and the basic research on the mechanism of thymosin drugs in anti-lung cancer are both systematically summarized and analyzed in this paper, along with future research directions.
Collapse
Affiliation(s)
| | - Jibin Lu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Faraoni P, Bellumori M, Cecchi L, Zonfrillo B, Innocenti M, Gnerucci A, Mulinacci N, Ranaldi F. AGS Gastric Cells: Antioxidant Activity and Metabolic Effects of Phenolic Extracts from Different Monocultivar Virgin Olive Oils. Antioxidants (Basel) 2023; 12:1347. [PMID: 37507887 PMCID: PMC10376124 DOI: 10.3390/antiox12071347] [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: 05/15/2023] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
The effects of the phenolic compounds of extra virgin olive oil (EVOO) on AGS cells have never been studied so far, which is the aim of this study. The profiles of the main phenolic components in EVOOs, mainly secoiridoid compounds derived from the transformation of oleuropein during the olive milling process, were evaluated and compared. Oils of different origins were evaluated aiming at verifying whether chemical differences in the phenolic composition of the dry extracts played a role in the metabolism and in maintaining the cellular redox state of AGS cells. The following key enzymes of some metabolic pathways were studied: lactate dehydrogenase, enolase, pyruvate kinase, glucose 6-phosphate dehydrogenase, citrate synthase, 3-Hydroxyacyl-CoA dehydrogenase and hexokinase. As confirmed through PCA analysis, pretreatments with the dry extracts of EVOOs at different concentrations appeared to be able to counteract the enzymatic activity alterations due to oxidative stress induced by H2O2 1 mM and 2 mM. The studied phytocomplexes showed the ability to protect AGS cells from oxidative damage and the secoiridoid derivatives from both oleuropein and ligstroside contributed to the observed effects. The results suggested that EVOOs with medium to high concentrations of phenols can exert this protection.
Collapse
Affiliation(s)
- Paola Faraoni
- Department of Experimental and Clinic Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139 Florence, FI, Italy
| | - Maria Bellumori
- Department of NEUROFARBA, Division of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via U. Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Lorenzo Cecchi
- Department of Agricultural, Food and Forestry Systems Management (DAGRI), University of Florence, Piazzale Delle Cascine 16, 50144 Florence, FI, Italy
| | - Beatrice Zonfrillo
- Department of NEUROFARBA, Division of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via U. Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Marzia Innocenti
- Department of NEUROFARBA, Division of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via U. Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Alessio Gnerucci
- Department of Physics and Astronomy, University of Florence, Via Sansone, 1, 50019 Sesto Fiorentino, FI, Italy
| | - Nadia Mulinacci
- Department of NEUROFARBA, Division of Pharmaceutical and Nutraceutical Sciences, University of Florence, Via U. Schiff 6, 50019 Sesto Fiorentino, FI, Italy
| | - Francesco Ranaldi
- Department of Experimental and Clinic Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139 Florence, FI, Italy
| |
Collapse
|
6
|
Wang S, Wei W, Yong H, Zhang Z, Zhang X, Zhang X, Wang S. Synergistic anti-cancer and attenuation effects of thymosin on chemotherapeutic drug vinorelbine in tumor-bearing zebrafish. Biomed Pharmacother 2023; 162:114633. [PMID: 37018994 DOI: 10.1016/j.biopha.2023.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/18/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Vinorelbine, the standard chemotherapy drug on advanced lung cancer, causes adverse events such as immunosuppression and bone marrow suppression. Thus, it is necessary to find drugs that could improve immune function and synergistically enhance the anti-tumor effect of vinorelbine. Thymosin is reported to inhibit tumor growth as an immunomodulator. Herein, to study the synergistic anti-cancer and attenuation effects of thymosin on vinorelbine, human lung cancer A549 cells that were labeled with CM-DiI were transplanted into zebrafish to establish the lung cancer xenotransplanted model. After treatment of vinorelbine and different concentrations of thymosin, the fluorescence intensity of CM-DiI-labeled A549 cells and the number of apoptotic muscle cells in the tumor-bearing zebrafish were detected. Besides, effects of thymosin on vinorelbine-reduced macrophages and T cells were identified in the transgenic zebrafish (Tg:zlyz-EGFP and Tg:rag2-DsRed). Then, the qRT-PCR was used to determine the alterations of the immune-related factors at the transcription level. Thymosin showed a marked synergistic anti-cancer effect with vinorelbine for the xenograft human lung cancer A549 cells, and the synergistic effect enhanced in a dose-dependent manner. Moreover, thymosin alleviated vinorelbine-induced muscle cell apoptosis, macrophage reduction, and T cell suppression. Compared with the vinorelbine group, co-administration with thymosin raised the mRNA levels of TNF-α, TNF-β, INF-γ, and GM-CSF. Thus, thymosin possesses synergistic anti-cancer effect on vinorelbine, and has protective effect on vinorelbine-induced immunosuppression. Thymosin, as an adjuvant immunomodulatory therapy, has great potential in enhancing the clinical application of vinorelbine.
Collapse
|
7
|
Lunin SM, Novoselova EG, Glushkova OV, Parfenyuk SB, Novoselova TV, Khrenov MO. Cell Senescence and Central Regulators of Immune Response. Int J Mol Sci 2022; 23:ijms23084109. [PMID: 35456927 PMCID: PMC9028919 DOI: 10.3390/ijms23084109] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/13/2022] Open
Abstract
Pathways regulating cell senescence and cell cycle underlie many processes associated with ageing and age-related pathologies, and they also mediate cellular responses to exposure to stressors. Meanwhile, there are central mechanisms of the regulation of stress responses that induce/enhance or weaken the response of the whole organism, such as hormones of the hypothalamic-pituitary-adrenal (HPA) axis, sympathetic and parasympathetic systems, thymic hormones, and the pineal hormone melatonin. Although there are many analyses considering relationships between the HPA axis and organism ageing, we found no systematic analyses of relationships between the neuroendocrine regulators of stress and inflammation and intracellular mechanisms controlling cell cycle, senescence, and apoptosis. Here, we provide a review of the effects of neuroendocrine regulators on these mechanisms. Our analysis allowed us to postulate a multilevel system of central regulators involving neurotransmitters, glucocorticoids, melatonin, and the thymic hormones. This system finely regulates the cell cycle and metabolic/catabolic processes depending on the level of systemic stress, stage of stress response, and energy capabilities of the body, shifting the balance between cell cycle progression, cell cycle stopping, senescence, and apoptosis. These processes and levels of regulation should be considered when studying the mechanisms of ageing and the proliferation on the level of the whole organism.
Collapse
|
8
|
Yang Z, Guo J, Cui K, Du Y, Zhao H, Zhu L, Weng L, Tang W, Guo J, Zhang T, Shi X, Zong H, Jin S, Ma W. Thymosin alpha-1 blocks the accumulation of myeloid suppressor cells in NSCLC by inhibiting VEGF production. Biomed Pharmacother 2020; 131:110740. [PMID: 32942159 DOI: 10.1016/j.biopha.2020.110740] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Thymosin alpha-1 (TA) has been reported to inhibit tumor growth as an immunomodulator. However, its mechanism of action in immunosuppressive cells is unclear. The purpose of this study was to investigate whether TA can reshape the immune microenvironment by inhibiting the function of myeloid-derived suppressor cells (MDSCs) in non-small cell lung carcinoma (NSCLC). METHODS The effects of TA on peripheral blood monocytic MDSCs (M-MDSCs) in patients with NSCLC and on the apoptosis and migration of M-MDSCs were studied. A mouse subcutaneous xenograft tumor model was constructed, and the effect of TA on M-MDSC migration was evaluated. Quantitative real-time PCR, Western blotting, flow cytometry and immunohistochemistry were used to examine the mechanism by which TA affects M-MDSCs. RESULTS TA not only promoted the apoptosis of M-MDSCs by reducing the Bcl-2/BAX ratio but also and more importantly inhibited the migration of MDSCs to the tumor microenvironment by suppressing the production of vascular endothelial growth factor (VEGF) through the downregulation of hypoxia-inducible factor (HIF)-1α in tumor cells. CONCLUSIONS TA may have a novel antitumor effect mediated by decreasing M-MDSC accumulation in the tumor microenvironment through reduced VEGF production.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiacheng Guo
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Kang Cui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yabing Du
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lili Zhu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lanling Weng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Wenxue Tang
- Center for Precision Medicine of Zhengzhou University, Zhengzhou, Henan, 450052, China; Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Jiancheng Guo
- Center for Precision Medicine of Zhengzhou University, Zhengzhou, Henan, 450052, China; Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Tengfei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaojing Shi
- Laboratory Animal Center, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hong Zong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuiling Jin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Wang Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
9
|
Costantini C, Bellet MM, Pariano M, Renga G, Stincardini C, Goldstein AL, Garaci E, Romani L. A Reappraisal of Thymosin Alpha1 in Cancer Therapy. Front Oncol 2019; 9:873. [PMID: 31555601 PMCID: PMC6742685 DOI: 10.3389/fonc.2019.00873] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Thymosin alpha1 (Tα1), an endogenous peptide first isolated from the thymic tissue in the mid-sixties, has gained considerable attention for its immunostimulatory activity that led to its application to diverse pathological conditions, including cancer. Studies in animal models and human patients have shown promising results in different types of malignancies, especially when Tα1 was used in combination with other chemo- and immune therapies. For this reason, the advancements in our knowledge on the adjuvant role of Tα1 have moved in parallel with the development of novel cancer therapies in a way that Tα1 was integrated to changing paradigms and protocols, and tested for increased efficacy and safety. Cancer immunotherapy has recently experienced a tremendous boost following the development and clinical application of immune checkpoint inhibitors. By unleashing the full potential of the adaptive immune response, checkpoint inhibitors were expected to be very effective against tumors, but it soon became clear that a widespread and successful application was not straightforward and shortcomings in efficacy and safety clearly emerged. This scenario led to the development of novel concepts in immunotherapy and the design of combination protocols to overcome these limitations, thus opening up novel opportunities for Tα1 application. Herein, we summarize in a historical perspective the use of Tα1 in cancer, with particular reference to melanoma, hepatocellular carcinoma and lung cancer. We will discuss the current limitations of checkpoint inhibitors in clinical practice and the mechanisms at the basis of a potential application of Tα1 in combination protocols.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Chen F, Zhang H, Jiang L, Wei W, Liu C, Cang S. Enhancing the cytotoxic efficacy of combined effect of doxorubicin and Cyclosporin encapsulated photoluminescent graphene dotted mesoporous nanoparticles against lung cancer cell-specific drug targeting for the nursing care of cancer patients. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 198:111578. [PMID: 31408842 DOI: 10.1016/j.jphotobiol.2019.111578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/14/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
In recent years, biological nanomedicine-based biomaterials have an extreme attention for biomedical uses, herein we examined a novel kind advance of photoluminescent Graphene quandum dots encapsulated mesoporous nanoparticles (GND@MSNs) encapsulated by well-known anticancer drugs Doxorubicin (DOX) and Cyclosporin (CsA) for lung carcinoma. Electron microscopic technique exhibit the nanostructure and spherical morphology of GND@MSNs+DOX+CsA with mean size ≈110 nm. Moreover, Dynamic Light Scattering (DLS) exposed that blended GND@MSNs+DOX+CsA nanoparticles were highly stable with extremely negatively charged nanoparticles. Raman investigation was done on the all naturally dynamic nanoparticles containing shed graphene to survey the blend condition of the graphene inside the silica mesoporous nanoparticles. GND@MSNs+DOX+CsA provided an outstanding anti-cancer efficiency against the lung cancer cell lines (i.e., A549 and HEL-299). MTT assay monitored that GND@MSNs, GND@MSNs+DOX and GND@MSNs+DOX+CsA have a robust toxicity behaviour on the A549 and HEL-299 model lung cancer cell lines. Additionally, investigation of the cell death was found on AO-EB, Hoechst 33452 staining and flowcytometry techniques. Furthermore, the DNA damage were confirmed by cell cycle arrest and comet assay. Hence, we suggesting that these GND@MSNs+DOX+CsA could be applied as auspicious drug vesicles for novel lung cancer therapeutic potential and new openings to solve the complexity of lung cancer in the care of cancer patients.
Collapse
Affiliation(s)
- Fengxia Chen
- Department of Oncology, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China
| | - Hongmei Zhang
- Department of Nursing Care, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China.
| | - Ling Jiang
- Department of Oncology, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China
| | - Wei Wei
- Department of Oncology, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China
| | - Chunchun Liu
- Department of Oncology, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou 450003, PR China.
| |
Collapse
|