1
|
Jeong Y, Shim YS, Jo YK, Cha HJ. Redox-activatable inhalable mucoadhesive proteinic nanotherapeutics for targeted treatment of lung cancer. Biomaterials 2025; 316:123004. [PMID: 39689461 DOI: 10.1016/j.biomaterials.2024.123004] [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: 06/27/2024] [Revised: 11/12/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Inhalation delivery has been considered a promising choice for treating lung cancer because it can shuttle therapeutic payloads directly to cancer tissues via simple and noninvasive procedures while reducing systemic toxicity. However, its clinical application still faces challenges, especially for delivering hydrophobic chemotherapeutic drugs, due to poor absorption on mucosal tissues and limited therapeutic performance. Herein, we propose inhalable mucoadhesive proteinic nanoparticles (NPs) capable of facilitating reliable pulmonary drug delivery and redox-responsive anticancer therapeutic effects to realize noninvasive, localized treatment of lung cancer in a highly biocompatible, site-specific manner. Thiolated mussel adhesive protein (MAP)-based NPs (thMAP NPs) can be administered to target tissues via an easy and facile nebulization process due to their superior MAP-driven adhesion ability and sufficient structural integrity. Curcumin (Cur)-loaded thMAP NPs (thMAP@Cur NPs) demonstrated efficient cellular uptake through the thiol-mediated pathway and controlled the intracellular release of Cur in response to the reductive environment in cancer cells. The nebulized thMAP@Cur NPs elicited prolonged retention in lung tissue without causing any detectable adverse effects, leading to significant inhibition of metastatic lung cancer in vivo. Thus, these protein-based redox-responsive mucoadhesive NPs hold great promise as robust inhalable drug delivery platforms to achieve effective, localized treatment of pulmonary cancer and other respiratory diseases.
Collapse
Affiliation(s)
- Yeonsu Jeong
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Yun Seop Shim
- Department of Biomedical Convergence Science and Technology, Advanced Institute of Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yun Kee Jo
- Department of Biomedical Convergence Science and Technology, Advanced Institute of Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea; Cell and Matrix Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea; Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
2
|
Hong WQ, Lee WH, Musa SH, Kamaruzaman NA, Loo CY. Evaluation of hydrogel loading with curcumin and silver nanoparticles: biocompatibilities and anti-biofilm activities. Biometals 2025; 38:663-682. [PMID: 39979667 DOI: 10.1007/s10534-025-00670-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Chronic wound healing is associated with prolonged elevated inflammation and high levels of oxidative stress leading to cell death. The majority of wounds are colonized with antibiotic-resistant bacterial biofilms such as Pseudomonas aeruginosa and Staphylococcus aureus. An ideal wound treatment should include agents with antioxidant, anti-inflammatory, and antibiofilm behavior. Therefore, in this study, a combination of curcumin nanoparticle (Cur-NP) and silver nanoparticle (AgNP) (Cur-NP/AgNP) loaded PVA hydrogel was used to inhibit the bacterial attachment and subsequent biofilm formation of P. aeruginosa and S. aureus. Cur was known for its antioxidant and anti-inflammatory effect while being non-toxic to cells. Meanwhile, AgNP demonstrated superior anti-bacterial and antibiofilm activities against both P. aeruginosa and S. aureus. Cur-NP/AgNP loaded PVA hydrogels completely inhibited the bacterial attachment and biofilm formation, possibly due to synergistic effect of Cur-NPs and AgNPs in killing the bacterial cells. It should be highlighted that no surviving bacterial cells were noted for Cur-NP/AgNP loaded hydrogels. On the other hand, AgNPs or Cur-NPs alone loaded hydrogels were unable to achieve complete inhibition of biofilm formation, even though significant reduction in the biofilm mass was noted compared with control samples. Cur-NP and AgNP exerted oxidative-stress induced cell death in HaCaT cells via mitochondrial dysfunction, mitochondrial membrane potential (MMP) reduction, adenosine triphosphate inhibition, and increased cytochrome C release. The toxicity of formulation followed the decreasing trend: Cur-NP/AgNP < AgNPs alone < Cur-NPs alone. Taken together, the combination of Cur-NP/AgNP completely inhibited bacterial biofilm formation through bactericidal effect on the planktonic cells while exerted the least toxic effects towards skin cells.
Collapse
Affiliation(s)
- Wei Qing Hong
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), 30450, Ipoh, Perak, Malaysia
| | - Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), 30450, Ipoh, Perak, Malaysia
| | - Siti Hajar Musa
- School of Pharmacy, Management and Science University, 40100, Shah Alam, Selangor Darul Ehsan, Malaysia
| | | | - Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), 30450, Ipoh, Perak, Malaysia.
| |
Collapse
|
3
|
Ullah R, Siraj M, Iqbal J, Abbasi BA. Potential of curcumin and its derivatives, modern insights on the anticancer properties: a comprehensive overview. Z NATURFORSCH C 2025:znc-2024-0220. [PMID: 40108840 DOI: 10.1515/znc-2024-0220] [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: 05/17/2024] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Globally, cancer is the top cause of mortality, placing a heavy load on the medical system. One of the first known secondary metabolites is curcumin, a bioactive substance. This study aims to emphasize the chemopreventive and chemotherapeutic properties of curcumin and its derivatives, therefore, offering important insights for the possible creation of certain supplemental medications for the treatment of different cancers. Electronic Google databases, including Google scholar, ResearchGate, PubMed/Medline, and ScienceDirect, were searched to gather pertinent data about the chemopreventive and chemotherapeutic effects of curcumin and its derivatives. Various studies have revealed a diverse array of significant biological effects. The majority of investigations pertaining to the potential anticancer effects and associated processes are currently in the experimental preclinical stage and lack sufficient clinical trial data to validate their findings. Clinical research is further needed to clarify the molecular processes and specific targeted action of curcumin and its derivatives, as well as their potential for toxicity and side effects in humans, in order to open up new therapeutic avenues for treating cancer.
Collapse
Affiliation(s)
- Rafi Ullah
- Department of Botany, Bacha Khan University, Charsadda 24420, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Siraj
- IBGE, University of Agriculture Peshawar, Peshawar, Pakistan
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda 24420, Khyber Pakhtunkhwa, Pakistan
| | - Banzeer Ahsan Abbasi
- Department of Botany, Rawalpindi Women University, 6th Road, Satellite Town, Rawalpindi, 46300, Pakistan
| |
Collapse
|
4
|
Kul Köprülü T, Balkan J, Gezer B, Erkal Çam B. Glycolytic pathway analysis and gene expression profiles of combination of aloe vera and paclitaxel on non-small cell lung cancer and breast cancer. Med Oncol 2024; 41:277. [PMID: 39400682 DOI: 10.1007/s12032-024-02506-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/14/2024] [Indexed: 10/15/2024]
Abstract
The purpose of this study is to enhance the effectiveness of known anticancer medications using natural compounds. The study investigated the impact of combining AVE with PAX on non-small cell lung cancer (A549) and breast cancer (MCF7). In this study, A549 and MCF7 cells were treated with PAX (5 μM), AVE (24 μg/mL), and a combination of PAX and AVE (5 μM + 24 μg/mL). The glucose consumption rates of the cells were determined by extracellular acidification rate (ECAR) thanks to the SeaHorse XFe24 instrument. In addition, gene expression profiles were determined by performing Total RNA sequencing with the Novaseq 6000 instrument. Finally, the expressions of GAPDH, BAX, and BCL-2 genes involved in the apoptotic pathway were detected by RT-qPCR. The combined application of PAX and AVE reduced the ECAR value in both cell lines. According to the RT-qPCR results, the expression level of the apoptotic gene BAX increased in both cell lines (p < 0.05). Total RNA sequencing revealed that the combination effects of PAX and AVE play a role in the ribosome mechanism, thereby affecting the protein translation system in MCF7 while apoptosis and cell cycle have come to the forefront in A549.
Collapse
Affiliation(s)
- Tuğba Kul Köprülü
- Experimental Medicine Application and Research Center, Validebağ Research Park, University of Health Sciences, Altunizade, Kalfaçeşme Street, Üsküdar, 34622, Istanbul, Turkey.
- Division of Medical Laboratory Techniques, Department of Medical Services and Techniques, University of Health Sciences, Istanbul, Turkey.
| | - Jülide Balkan
- Department of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, 34622, Istanbul, Turkey
| | - Bahar Gezer
- Department of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, 34622, Istanbul, Turkey
| | - Burçin Erkal Çam
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Yıldız Technical University, Esenler, Istanbul, Turkey
| |
Collapse
|
5
|
Yong J, Shu H, Zhang X, Yang K, Luo G, Yu L, Li J, Huang H. Natural Products-Based Inhaled Formulations for Treating Pulmonary Diseases. Int J Nanomedicine 2024; 19:1723-1748. [PMID: 38414528 PMCID: PMC10898359 DOI: 10.2147/ijn.s451206] [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: 11/22/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Given the unique physiological and pathological characteristics of the lung, the direct, inhalable route is more conducive to pulmonary drug delivery and disease control than traditional systemic drug delivery, significantly circumventing drug loss, off-target effects, systemic and organ toxicity, etc., and is widely regarded as the preferred regimen for pulmonary drug delivery. However, very few lung diseases are currently treated with the preferred inhaled formulations, such as asthma, chronic obstructive pulmonary disease and pulmonary hypertension. And there is a lack of appropriate inhaled formulations for other critical lung diseases, such as lung cancer and pulmonary fibrosis, due to the fact that the physicochemical properties of the drugs and their pharmacokinetic profiles do not match the physiology of the lung, and conventional inhalation devices are unable to deliver them to the specific parts of the lung. Phytochemicals of natural origin, due to their wide availability and clear safety profile, hold great promise for the preparation of inhalable formulations to improve the current dilemma in the treatment of lung diseases. In particular, the preparation of inhalable formulations based on nano- and microparticulate carriers for drug delivery to deep lung tissues, which overcome the shortcomings of conventional inhalation therapies while targeting the drug activity directly to a specific part of the lung, may be the best approach to change the current dilemma of lung disease treatment. In this review, we discuss recent advances in nano- and micron-carrier-based inhalation formulations for the delivery of natural products for the treatment of pulmonary diseases, which may represent an opportunity for practical clinical translation of natural products.
Collapse
Affiliation(s)
- Jiangyan Yong
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Hongli Shu
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, People’s Republic of China
| | - Xiao Zhang
- Department of Clinical Laboratory, Chengdu Children Special Hospital, Chengdu, Sichuan, 610031, People’s Republic of China
| | - Kun Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Guining Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Lu Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Jiaqi Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People’s Republic of China
| | - Hong Huang
- Department of Clinical Laboratory, the People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, 401121, People’s Republic of China
| |
Collapse
|
6
|
Hu Y, Cheng L, Du S, Wang K, Liu S. Antioxidant curcumin induces oxidative stress to kill tumor cells (Review). Oncol Lett 2024; 27:67. [PMID: 38192657 PMCID: PMC10773205 DOI: 10.3892/ol.2023.14200] [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: 07/15/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
Curcumin is a plant polyphenol in turmeric root and a potent antioxidant. It binds to antioxidant response elements for gene regulation by nuclear factor erythroid 2-related factor 2, thereby suppressing reactive oxygen species (ROS) and exerting anti-inflammatory, anti-infective and other pharmacological effects. Of note, curcumin induces oxidative stress in tumors. It binds to several enzymes in tumors, such as carbonyl reductases, glutathione S-transferase P1 and nicotinamide adenine dinucleotide phosphate to induce mitochondrial damage, increase ROS production and ultimately induce tumor cell death. However, the instability and poor pharmacokinetic profile of curcumin in vivo limit its clinical application. Therefore, the effects of curcumin in vivo may be enhanced through its combination with drugs, derivative development and nanocarriers. In the present review, the mechanisms of curcumin that induce tumor cell death through oxidative stress are discussed. In addition, the methods used to enhance the antitumor activity of curcumin are described. Finally, the existing knowledge on the functions of curcumin in tumors, particularly in terms of oxidative stress, are summarized to facilitate future curcumin research.
Collapse
Affiliation(s)
- Ye Hu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Lei Cheng
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Shuguang Du
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Kesi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| |
Collapse
|
7
|
Gao G, Zhou W, Jiang X, Ma J. Bovine serum albumin and folic acid-modified aurum nanoparticles loaded with paclitaxel and curcumin enhance radiotherapy sensitization for esophageal cancer. Int J Radiat Biol 2023; 100:411-419. [PMID: 37934908 DOI: 10.1080/09553002.2023.2281524] [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: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Nanocarrier systems have been used in the study of esophageal cancer (EC) and other diseases, with significant advantages in improving the non-targeted and nonspecific toxicity of traditional formulations. Some chemotherapeutic drugs and high atomic number nanomaterials have sensitization effects on ionizing radiation and can be used as chemoradiation sensitizers. METHODS Aurum (Au) nanoparticles were modified by bovine serum albumin (BSA) and folic acid (FA), and were core-loaded with paclitaxel (PTX) and curcumin (CUR). The basic characteristics of FA-BSA-Au@PTX/CUR nanomedicines were evaluated by transmission electron microscopy, Fourier transform infrared spectroscopy, and Malvern Zetasizer. The encapsulation and release of drugs were monitored by ultraviolet-visible spectrophotometry (UV-Vis). The biological toxicity and radiotherapy sensitization effect of FA-BSA-Au@PTX/CUR were observed by cell viability, colony formation, cell apoptosis, cell cycle distribution, and γ-H2AX analysis experiments. RESULTS The prepared nanomedicines showed good stability and spherical morphology. The results of cell uptake and cell viability detection revealed that FA-BSA-Au@PTX/CUR could specifically target EC cell KYSE150 and exert a certain inhibitory effect on proliferation, with no obvious toxicity on healthy cells Het-1A. In addition, the results of the colony formation experiment, cell apoptosis detection, cell cycle distribution, and γ-H2AX analysis showed that compared with X-rays alone, FA-BSA-Au@PTX/CUR combined with X-rays exhibited relatively stronger radiotherapy sensitization and anti-tumor activity. CONCLUSIONS FA-BSA-Au@PTX/CUR could target EC cancer cells and act as a safe and effective radiotherapy sensitizer to improve the radiotherapy efficacy of EC.
Collapse
Affiliation(s)
- Guangyi Gao
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Wenhang Zhou
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Xuan Jiang
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, China
| | - Jun Ma
- Department of Oncology, Huai'an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai'an, China
| |
Collapse
|
8
|
Tan J, Zhuo Z, Wang X, Zhang Y, Qian Y, Lin F. Secretome of EMSCs neutralizes LPS‑induced acute lung injury via aerosol administration. Int J Mol Med 2023; 52:104. [PMID: 37772372 PMCID: PMC10558219 DOI: 10.3892/ijmm.2023.5307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Ectodermal mesenchymal stem cells (EMSCs) are cells harvested from the stem cell niche (nasal mucosa) with high therapeutic potential. To the best of our knowledge, however, the anti‑inflammatory properties of these neural crest‑derived EMSCs have been rarely reported. The present study aimed to explore the effects of aerosolized EMSC‑Secretome (EMSC‑Sec) and clarify underlying mechanisms in treating acute lung injury (ALI). EMSCs were isolated by adherent method and identified by immunofluorescence staining. EMSC‑Sec was collected and evaluated using western blotting, BCA and ELISA tests. Then, mouse lung epithelial cells (MLE‑12) were used to mimic inflammatory stimulation with lipopolysaccharide (LPS). After developing an ALI model through intraperitoneal injection of LPS, mice were treated with an EMSC‑Sec spray. The lung in each group underwent an observation and measurement to preliminarily assess the extent of damage. H&E staining, immunohistochemical staining, immunofluorescence and western‑blotting were utilized to further access the impacts of EMSC‑Sec. The results showed that EMSC‑Sec had great anti‑inflammatory potential and was highly successful in vitro and in vivo. EMSC‑Sec mitigated LPS‑induced ALI with low inflammatory cell inflation and mild damage. EMSC‑Sec could regulate inflammation via the NF‑κB(p50/p65)/NLRP3 pathway. Overall, the present study demonstrated that EMSC‑Sec regulated inflammation, hoping to provide a novel strategy for ALI treatment.
Collapse
Affiliation(s)
- Jianing Tan
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Ziliang Zhuo
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Xiuyu Wang
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Yanshuang Zhang
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Yucheng Qian
- Department of Neurology, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University, Changshu, Suzhou 215500, P.R. China
| | - Fangfang Lin
- Department of Oncology, The First People's Hospital of Zhenjiang, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| |
Collapse
|
9
|
Dytrych P, Kejík Z, Hajduch J, Kaplánek R, Veselá K, Kučnirová K, Skaličková M, Venhauerová A, Hoskovec D, Martásek P, Jakubek M. Therapeutic potential and limitations of curcumin as antimetastatic agent. Biomed Pharmacother 2023; 163:114758. [PMID: 37141738 DOI: 10.1016/j.biopha.2023.114758] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023] Open
Abstract
Treatment of metastatic cancer is one of the biggest challenges in anticancer therapy. Curcumin is interesting nature polyphenolic compound with unique biological and medicinal effects, including repression of metastases. High impact studies imply that curcumin can modulate the immune system, independently target various metastatic signalling pathways, and repress migration and invasiveness of cancer cells. This review discusses the potential of curcumin as an antimetastatic agent and describes potential mechanisms of its antimetastatic activity. In addition, possible strategies (curcumin formulation, optimization of the method of administration and modification of its structure motif) to overcome its limitation such as low solubility and bioactivity are also presented. These strategies are discussed in the context of clinical trials and relevant biological studies.
Collapse
Affiliation(s)
- Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, 121 08 Prague, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic
| | - Kateřina Kučnirová
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic
| | - Anna Venhauerová
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, 121 08 Prague, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic.
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 128 08 Prague, Czech Republic.
| |
Collapse
|
10
|
Jakobušić Brala C, Karković Marković A, Kugić A, Torić J, Barbarić M. Combination Chemotherapy with Selected Polyphenols in Preclinical and Clinical Studies-An Update Overview. Molecules 2023; 28:molecules28093746. [PMID: 37175156 PMCID: PMC10180288 DOI: 10.3390/molecules28093746] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
This review article describes studies published over the past five years on the combination of polyphenols, which are the most studied in the field of anticancer effects (curcumin, quercetin, resveratrol, epigallocatechin gallate, and apigenin) and chemotherapeutics such as cisplatin, 5-fluorouracil, oxaliplatin, paclitaxel, etc. According to WHO data, research has been limited to five cancers with the highest morbidity rate (lung, colorectal, liver, gastric, and breast cancer). A systematic review of articles published in the past five years (from January 2018 to January 2023) was carried out with the help of all Web of Science databases and the available base of clinical studies. Based on the preclinical studies presented in this review, polyphenols can enhance drug efficacy and reduce chemoresistance through different molecular mechanisms. Considering the large number of studies, curcumin could be a molecule in future chemotherapy cocktails. One of the main problems in clinical research is related to the limited bioavailability of most polyphenols. The design of a new co-delivery system for drugs and polyphenols is essential for future clinical research. Some polyphenols work in synergy with chemotherapeutic drugs, but some polyphenols can act antagonistically, so caution is always required.
Collapse
Affiliation(s)
- Cvijeta Jakobušić Brala
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Ana Karković Marković
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Azra Kugić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Jelena Torić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| | - Monika Barbarić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10 000 Zagreb, Croatia
| |
Collapse
|
11
|
Pulmonary delivery of curcumin and quercetin nanoparticles for lung cancer – Part 2: Toxicity and endocytosis. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
12
|
Nie Q, Wang C, Xu H, Mittal P, Naeem A, Zhou P, Li H, Zhang Y, Guo T, Sun L, Zhang J. Highly efficient pulmonary delivery of levo-tetrahydropalmatine using γ-cyclodextrin metal-organic framework as a drug delivery platform. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
13
|
Pi C, Zhao W, Zeng M, Yuan J, Shen H, Li K, Su Z, Liu Z, Wen J, Song X, Lee RJ, Wei Y, Zhao L. Anti-lung cancer effect of paclitaxel solid lipid nanoparticles delivery system with curcumin as co-loading partner in vitro and in vivo. Drug Deliv 2022; 29:1878-1891. [PMID: 35748365 PMCID: PMC9246235 DOI: 10.1080/10717544.2022.2086938] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The main aim of this study was to improve the therapeutic potential of a paclitaxel (PTX) and curcumin (CU) combination regimen using solid lipid nanoparticles (SLNs). PTX and CU were successfully co-encapsulated at a predetermined ratio in SLNs (PC-SLNs) with high encapsulation efficiency (CU: 97.6%, PTX: 95.8%), appropriate particle size (121.8 ± 1.69 nm), small PDI (0.267 ± 0.023), and negative zeta potential (–30.4 ± 1.25 mV). Compared with PTX or the combination of CU and PTX (CU + PTX), PC-SLNs can greatly reduce the dose of PTX while still achieving the same therapeutic effect on four cancer cell lines, among which the inhibitory effect on A549 lung cancer cells was the strongest. PC-SLNs improved the area under the curve (CU: 1.40-fold; PTX: 2.88-fold), prolonged the residence time (CU: 6.94-fold; PTX: 2.51-fold), and increased the half-life (CU: 5.62-fold; PTX: 6.46-fold), achieving long circulation. PC-SLNs were used to treat lung cancer in a nude mouse xenograft tumor model and the tumor suppression rate reached 78.42%, while those of PTX and (CU + PTX) were 40.53% and 51.56%, respectively. As PC-SLNs can prevent P-glycoprotein efflux, reverse MDR and downregulate the NF-κB pathway. PC-SLNs are a potential antineoplastic agent that is more effective and less toxic in treating lung cancer.
Collapse
Affiliation(s)
- Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Wenmei Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Mingtang Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Jiyuan Yuan
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R China
| | - Hongping Shen
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R China
| | - Ke Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Zhilian Su
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd, Luzhou, Sichuan, P. R. China.,Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, Shapingba, P. R. China
| | - Jie Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Xinjie Song
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang, P. R. China.,Department of Food Science and Technology, Yeungnam University, Gyeongsan-si, Republic of Korea
| | - Robert J Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, P. R China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China.,Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P. R. China
| |
Collapse
|
14
|
Adhikari BR, Dummer J, Gordon KC, Das SC. An expert opinion on respiratory delivery of high dose powders for lung infections. Expert Opin Drug Deliv 2022; 19:795-813. [PMID: 35695722 DOI: 10.1080/17425247.2022.2089111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION High dose powder inhalation is evolving as an important approach to to treat lung infections. It is important to its identify applications, consider the factors affecting high dose powder delivery, and assess the effect of high dose drugs in patients. AREA COVERED Both current and pipeline high dose inhalers and their applications have been summarized. Challenges and opportunities to high dose delivery have been highlighted after reviewing formulation techniques in the context of factors affecting aerosolization, devices, and patient factors. EXPERT OPINION High dose inhaled delivery of antimicrobials is an innovative way to increase treatment efficacy of respiratory infections, tackle drug resistance, and the scarcity of new antimicrobials. The high dose inhaled technology also has potential for systemic action; however, innovations in formulation strategies and devices are required to realize its full potential. Advances in formulation strategies include the use of excipients or the engineering of particles to decrease the cohesive property of microparticles and their packing density. Similarly, selection of a synergistic drug instead of an excipient can be considered to increase aerosolization and stability. Device development focused on improving dispersion and loading capacity is also important, and modification of existing devices for high dose delivery can also be considered.
Collapse
Affiliation(s)
| | - Jack Dummer
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Keith C Gordon
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Saddiq AA, El-Far AH, Mohamed Abdullah SA, Godugu K, Almaghrabi OA, Mousa SA. Curcumin, thymoquinone, and 3, 3′-diindolylmethane combinations attenuate lung and liver cancers progression. Front Pharmacol 2022; 13:936996. [PMID: 35847018 PMCID: PMC9277483 DOI: 10.3389/fphar.2022.936996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022] Open
Abstract
Cancer can develop due to abnormal cell proliferation in any body’s cells, so there are over a hundred different types of cancer, each with its distinct behavior and response to treatment. Therefore, many studies have been conducted to slow cancer progression and find effective and safe therapies. Nutraceuticals have great attention for their anticancer potential. Therefore, the current study was conducted to investigate the anticancer effects of curcumin (Cur), thymoquinone (TQ), and 3, 3′-diindolylmethane (DIM) combinations on lung (A549) and liver (HepG2) cancer cell lines’ progression. Results showed that triple (Cur + TQ + DIM) and double (Cur + TQ, Cur + DIM, and TQ + DIM) combinations of Cur, TQ, and DIM significantly increased apoptosis with elevation of caspase-3 protein levels. Also, these combinations exhibited significantly decreased cell proliferation, migration, colony formation activities, phosphatidylinositol 3-kinase (PI3K), and protein kinase B (AKT) protein levels with S phase reduction. Triple and double combinations of Cur, TQ, and DIM hindered tumor weight and angiogenesis of A549 and HepG2 implants in the chorioallantoic membrane model. Interestingly, Cur, TQ, and DIM combinations are considered promising for suppressing cancer progression via inhibiting tumor angiogenesis. Further preclinical and clinical investigations are warranted.
Collapse
Affiliation(s)
- Amna A. Saddiq
- College of Sciences, Department of Biology, University of Jeddah, Jeddah, Saudi Arabia
| | - Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
- *Correspondence: Ali H. El-Far,
| | - Shymaa Abdullah Mohamed Abdullah
- Molecular Biology Unit, Medical Technology Center and Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, United States
| | - Omar A. Almaghrabi
- College of Sciences, Department of Biology, University of Jeddah, Jeddah, Saudi Arabia
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, United States
| |
Collapse
|
16
|
Toxicity of curcumin nanoparticles towards alveolar macrophage: Effects of surface charges. Food Chem Toxicol 2022; 163:112976. [PMID: 35364129 DOI: 10.1016/j.fct.2022.112976] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 12/21/2022]
Abstract
Curcumin has been used for chronic lung diseases management due to its diversified molecular actions. However, the potential cytotoxicity which occurs in cells following the exposure to high concentrations of curcumin has been overlooked. This study evaluated the toxic events of curcumin nanoparticles (Cur-NPs) with alterable surface polarity in alveolar macrophages (NR8383). We aimed to establish the correlation between the toxicity of Cur-NPs with different surface charges and the internalization mechanisms of the NPs. Toxicity data showed that positively charged Cur-NPs (IC50: 9.77 ± 0.5 μg/mL) was the most potent against NR8383, followed by negatively charged Cur-NPs (IC50:13.33 ± 0.9 μg/mL) and neutral Cur-NPs (IC50:18.68 ± 1.2 μg/mL). Results from mitochondrial membrane potential, ATP content and intracellular ROS in NR8383 showed similar ranking to the toxicity assay. The predominant uptake pathway for positively and negatively charged Cur-NPs was via clathrin-mediated endocytosis, while neutral Cur-NPs was internalized via phagocytosis, micropinocytosis and clathrin-mediated endocytosis. Positively charged Cur-NPs mediates the cytotoxicity of NR8383 via lysosomal and mitochondrial-associated destabilization upon entry. In conclusion, the cytotoxicity of Cur-NPs on NR8383 is surface-charge dependent, which in turn is associated to the uptake pathway and localization of Cur-NPs in cells.
Collapse
|
17
|
Vemuri SK, Halder S, Banala RR, Rachamalla HK, Devraj VM, Mallarpu CS, Neerudu UK, Bodlapati R, Mukherjee S, Venkata SGP, Venkata GRA, Thakkumalai M, Jana K. Modulatory Effects of Biosynthesized Gold Nanoparticles Conjugated with Curcumin and Paclitaxel on Tumorigenesis and Metastatic Pathways-In Vitro and In Vivo Studies. Int J Mol Sci 2022; 23:ijms23042150. [PMID: 35216264 PMCID: PMC8876049 DOI: 10.3390/ijms23042150] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023] Open
Abstract
Background: Breast cancer is the most common cancer in women globally, and diagnosing it early and finding potential drug candidates against multi-drug resistant metastatic breast cancers provide the possibilities of better treatment and extending life. Methods: The current study aimed to evaluate the synergistic anti-metastatic activity of Curcumin (Cur) and Paclitaxel (Pacli) individually, the combination of Curcumin–Paclitaxel (CP), and also in conjugation with gold nanoparticles (AuNP–Curcumin (Au-C), AuNP–Paclitaxel (Au-P), and AuNP–Curcumin–Paclitaxel (Au-CP)) in various in vitro and in vivo models. Results: The results from combination treatments of CP and Au-CP demonstrated excellent synergistic cytotoxic effects in triple-negative breast cancer cell lines (MDA MB 231 and 4T1) in in vitro and in vivo mouse models. Detailed mechanistic studies were performed that reveal that the anti-cancer effects were associated with the downregulation of the expression of VEGF, CYCLIN-D1, and STAT-3 genes and upregulation of the apoptotic Caspase-9 gene. The group of mice that received CP combination therapy (with and without gold nanoparticles) showed a significant reduction in the size of tumor when compared to the Pacli alone treatment and control groups. Conclusions: Together, the results suggest that the delivery of gold conjugated Au-CP formulations may help in modulating the outcomes of chemotherapy. The present study is well supported with observations from cell-based assays, molecular and histopathological analyses.
Collapse
Affiliation(s)
- Satish Kumar Vemuri
- Sunshine Medical Academy Research and Technoloy (SMART), Sunshine Hospitals, PG Road, Secunderabad 500003, Telangana, India; (R.R.B.); (V.M.D.); (S.G.P.V.); (G.R.A.V.)
- Department of Biochemistry, Bharathidasan University Constituent College for Women, Tiruchirappalli 620009, Tamil Nadu, India;
- Correspondence: (S.K.V.); (K.J.); Tel.: +91-807-431-7348 (S.K.V.); +91-900-704-2850 (K.J.)
| | - Satyajit Halder
- Division of Molecular Medicine, Centenary Campus, Bose Institute, P-1/12 C.I.T. Scheme VII-M, Kolkata 700054, West Bengal, India;
| | - Rajkiran Reddy Banala
- Sunshine Medical Academy Research and Technoloy (SMART), Sunshine Hospitals, PG Road, Secunderabad 500003, Telangana, India; (R.R.B.); (V.M.D.); (S.G.P.V.); (G.R.A.V.)
| | - Hari Krishnreddy Rachamalla
- Biomaterials Group, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad 500007, Telangana, India;
| | - Vijaya Madhuri Devraj
- Sunshine Medical Academy Research and Technoloy (SMART), Sunshine Hospitals, PG Road, Secunderabad 500003, Telangana, India; (R.R.B.); (V.M.D.); (S.G.P.V.); (G.R.A.V.)
| | | | - Uttam Kumar Neerudu
- Department of Biochemistry, Osmania University, Hyderabad 500007, Telangana, India;
| | - Ravikiran Bodlapati
- TBRC, Business Research Private Limited, Hyderabad 500033, Telangana, India;
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX 77030, USA;
| | - Subbaiah Goli Peda Venkata
- Sunshine Medical Academy Research and Technoloy (SMART), Sunshine Hospitals, PG Road, Secunderabad 500003, Telangana, India; (R.R.B.); (V.M.D.); (S.G.P.V.); (G.R.A.V.)
| | - Gurava Reddy Annapareddy Venkata
- Sunshine Medical Academy Research and Technoloy (SMART), Sunshine Hospitals, PG Road, Secunderabad 500003, Telangana, India; (R.R.B.); (V.M.D.); (S.G.P.V.); (G.R.A.V.)
| | - Malarvilli Thakkumalai
- Department of Biochemistry, Bharathidasan University Constituent College for Women, Tiruchirappalli 620009, Tamil Nadu, India;
| | - Kuladip Jana
- Division of Molecular Medicine, Centenary Campus, Bose Institute, P-1/12 C.I.T. Scheme VII-M, Kolkata 700054, West Bengal, India;
- Correspondence: (S.K.V.); (K.J.); Tel.: +91-807-431-7348 (S.K.V.); +91-900-704-2850 (K.J.)
| |
Collapse
|
18
|
Design and Characterization of Atorvastatin Dry Powder Formulation as a potential Lung Cancer Treatment. Saudi Pharm J 2022; 29:1449-1457. [PMID: 35002383 PMCID: PMC8720807 DOI: 10.1016/j.jsps.2021.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer death. Many studies have shown the beneficial effects of Atorvastatin in decreasing the mortality risk and improving survival among patients with lung cancer. This research paper focuses on improving AVT cytotoxic activity and cellular uptake by developing mannitol microcarriers as a promising drug delivery system for lung cancer treatment and, studying the impact of improving inhalation deposition on the delivery and Dry Powder formulations efficiency. The AVT loaded mannitol (AM) microparticles (AVT-AM) formulation was prepared by spray drying and characterized for its physicochemical properties and aerodynamic deposition. The results revealed that the AVT-AM formulation has good flow properties and aerosol deposition with a particle size of 3418 nm ± 26.86. The formulation was also assessed in vitro for cytotoxicity effects (proliferation, apoptosis, and cell cycle progression) on A549 human lung adenocarcinoma. Compared with free AVT, the AVT-AM formulation has significantly higher cellular uptake and anti-cancer properties by disrupting cell cycle progression via either apoptosis or cell cycle arrest in the G2/M phase. This study shows that AVT loaded mannitol microcarriers may provide a potentially effective and sustained pulmonary drug delivery for lung cancer treatment.
Collapse
|
19
|
Bedford R, Perkins E, Clements J, Hollings M. Recent advancements and application of in vitro models for predicting inhalation toxicity in humans. Toxicol In Vitro 2021; 79:105299. [PMID: 34920082 DOI: 10.1016/j.tiv.2021.105299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/20/2021] [Accepted: 12/10/2021] [Indexed: 12/01/2022]
Abstract
Animals have been indispensable in testing chemicals that can pose a risk to human health, including those delivered by inhalation. In recent years, the combination of societal debate on the use of animals in research and testing, the drive to continually enhance testing methodologies, and technology advancements have prompted a range of initiatives to develop non-animal alternative approaches for toxicity testing. In this review, we discuss emerging in vitro techniques being developed for the testing of inhaled compounds. Advanced tissue models that are able to recreate the human response to toxic exposures alongside examples of their ability to complement in vivo techniques are described. Furthermore, technology being developed that can provide multi-organ toxicity assessments are discussed.
Collapse
Affiliation(s)
- R Bedford
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - E Perkins
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - J Clements
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - M Hollings
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| |
Collapse
|
20
|
Latest Innovations and Nanotechnologies with Curcumin as a Nature-Inspired Photosensitizer Applied in the Photodynamic Therapy of Cancer. Pharmaceutics 2021; 13:pharmaceutics13101562. [PMID: 34683855 PMCID: PMC8539945 DOI: 10.3390/pharmaceutics13101562] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/27/2022] Open
Abstract
In the context of the high incidence of cancer worldwide, state-of-the-art photodynamic therapy (PDT) has entered as a usual protocol of attempting to eradicate cancer as a minimally invasive procedure, along with pharmacological resources and radiation therapy. The photosensitizer (PS) excited at certain wavelengths of the applied light source, in the presence of oxygen releases several free radicals and various oxidation products with high cytotoxic potential, which will lead to cell death in irradiated cancerous tissues. Current research focuses on the potential of natural products as a superior generation of photosensitizers, which through the latest nanotechnologies target tumors better, are less toxic to neighboring tissues, but at the same time, have improved light absorption for the more aggressive and widespread forms of cancer. Curcumin incorporated into nanotechnologies has a higher intracellular absorption, a higher targeting rate, increased toxicity to tumor cells, accelerates the activity of caspases and DNA cleavage, decreases the mitochondrial activity of cancer cells, decreases their viability and proliferation, decreases angiogenesis, and finally induces apoptosis. It reduces the size of the primary tumor, reverses multidrug resistance in chemotherapy and decreases resistance to radiation therapy in neoplasms. Current research has shown that the use of PDT and nanoformulations of curcumin has a modulating effect on ROS generation, so light or laser irradiation will lead to excessive ROS growth, while nanocurcumin will reduce the activation of ROS-producing enzymes or will determine the quick removal of ROS, seemingly opposite but synergistic phenomena by inducing neoplasm apoptosis, but at the same time, accelerating the repair of nearby tissue. The latest curcumin nanoformulations have a huge potential to optimize PDT, to overcome major side effects, resistance to chemotherapy, relapses and metastases. All the studies reviewed and presented revealed great potential for the applicability of nanoformulations of curcumin and PDT in cancer therapy.
Collapse
|
21
|
Abdulbaqi IM, Assi RA, Yaghmur A, Darwis Y, Mohtar N, Parumasivam T, Saqallah FG, Wahab HA. Pulmonary Delivery of Anticancer Drugs via Lipid-Based Nanocarriers for the Treatment of Lung Cancer: An Update. Pharmaceuticals (Basel) 2021; 14:725. [PMID: 34451824 PMCID: PMC8400724 DOI: 10.3390/ph14080725] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths, responsible for approximately 18.4% of all cancer mortalities in both sexes combined. The use of systemic therapeutics remains one of the primary treatments for LC. However, the therapeutic efficacy of these agents is limited due to their associated severe adverse effects, systemic toxicity and poor selectivity. In contrast, pulmonary delivery of anticancer drugs can provide many advantages over conventional routes. The inhalation route allows the direct delivery of chemotherapeutic agents to the target LC cells with high local concertation that may enhance the antitumor activity and lead to lower dosing and fewer systemic toxicities. Nevertheless, this route faces by many physiological barriers and technological challenges that may significantly affect the lung deposition, retention, and efficacy of anticancer drugs. The use of lipid-based nanocarriers could potentially overcome these problems owing to their unique characteristics, such as the ability to entrap drugs with various physicochemical properties, and their enhanced permeability and retention (EPR) effect for passive targeting. Besides, they can be functionalized with different targeting moieties for active targeting. This article highlights the physiological, physicochemical, and technological considerations for efficient inhalable anticancer delivery using lipid-based nanocarriers and their cutting-edge role in LC treatment.
Collapse
Affiliation(s)
- Ibrahim M. Abdulbaqi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
- College of Pharmacy, Al-Kitab University, Altun kupri, Kirkuk 36001, Iraq
| | - Reem Abou Assi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
- College of Pharmacy, Al-Kitab University, Altun kupri, Kirkuk 36001, Iraq
| | - Anan Yaghmur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark;
| | - Yusrida Darwis
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
| | - Noratiqah Mohtar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
| | - Thaigarajan Parumasivam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
| | - Fadi G. Saqallah
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (I.M.A.); (R.A.A.); (N.M.); (T.P.); (F.G.S.)
| |
Collapse
|
22
|
Wu Q, Ou H, Shang Y, Zhang X, Wu J, Fan F. Nanoscale Formulations: Incorporating Curcumin into Combination Strategies for the Treatment of Lung Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2695-2709. [PMID: 34188448 PMCID: PMC8232383 DOI: 10.2147/dddt.s311107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer remains the most common cancer worldwide. Although significant advances in screening have been made and early diagnosis strategies and therapeutic regimens have been developed, the overall survival rate remains bleak. Curcumin is extracted from the rhizomes of turmeric and exhibits a wide range of biological activities. In lung cancer, evidence has shown that curcumin can markedly inhibit tumor growth, invasion and metastasis, overcome resistance to therapy, and even eliminate cancer stem cells (CSCs). Herein, the underlying molecular mechanisms of curcumin were summarized by distinct biological processes. To solve the limiting factors that curtail the clinical applications of curcumin, nanoformulations encapsulating curcumin were surveyed in detail. Nanoparticles, including liposomes, micelles, carbon nanotubes (CNTs), solid lipid nanoparticles (SLNs), nanosuspensions, and nanoemulsions, were explored as proper carriers of curcumin. Moreover, it was firmly verified that curcumin has the ability to sensitize lung cancer cells to chemotherapeutic drugs, such as cisplatin and docetaxel, and to various targeted therapies. Regarding the advantages and drawbacks of curcumin, we concluded that combination therapy based on nanoparticles would be the optimal approach to broaden the application of curcumin in the clinic in the near future.
Collapse
Affiliation(s)
- Quhui Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Huiping Ou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Yan Shang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Xi Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Junyong Wu
- Department of Pharmacy, Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Fuyuan Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
23
|
Sun J, Guo Y, Fan Y, Wang Q, Zhang Q, Lai D. Decreased expression of IDH1 by chronic unpredictable stress suppresses proliferation and accelerates senescence of granulosa cells through ROS activated MAPK signaling pathways. Free Radic Biol Med 2021; 169:122-136. [PMID: 33865962 DOI: 10.1016/j.freeradbiomed.2021.04.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/20/2022]
Abstract
Studies suggested that psychosocial stress was associated with female fertility decline, but the underlying mechanisms remained unclear. Granulosa cells (GCs) are important somatic cells to support follicular development and oocyte maturation. Herein, by using a mouse model of chronic unpredictable stress (CUS), we found that CUS induced oxidative stress damage in mouse ovaries, also inhibited GCs proliferation and accelerated GCs senescence. Isocitrate dehydrogenase-1 (IDH1), an antioxidant related gene by generating NADPH, was shown to be downregulated in GCs of CUS mice. Consistently, IDH1 knockdown inhibited cell proliferation and accelerated cellular senescence in KGN cells in vitro. In addition, IDH1 knockdown increased ROS content, induced autophagy activation and triggered cell cycle arrest in S and G2/M phases in KGN cells, which could be rescued by N-acetyl-l-cysteine (NAC), a ROS scavenger in these cells. Besides, IDH1 knockdown activated MAPK signaling pathways, including ERK, JNK and p38 signaling pathways in KGN cells, while NAC could suppress the activation. Through using inhibitors of MAPK signaling pathways, we showed that the activation of ERK pathway participated in autophagy related cell proliferation inhibition and cellular senescence, whereas JNK and p38 MAPK signaling pathways took part in regulation cell cycle arrest associated cell proliferation inhibitory and senescence in IDH1 knockdown KGN cells. Our findings suggested that downregulated expression of IDH1 induced by CUS has a physiological function in GCs proliferation and senescence through ROS activated MAPK signaling pathways, and improvement of IDH1 activity might be a beneficial therapeutic strategy for ovarian dysfunction.
Collapse
Affiliation(s)
- Junyan Sun
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China
| | - Ying Guo
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China
| | - Yihui Fan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China
| | - Qian Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China; Shanghai Municipal Key Clinical Speciality, Shanghai, 200030, China.
| |
Collapse
|
24
|
Hemmati K, Ahmadi Nasab N, Hesaraki S, Nezafati N. In vitro evaluation of curcumin-loaded chitosan-coated hydroxyapatite nanocarriers as a potential system for effective treatment of cancer. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1267-1287. [PMID: 33820489 DOI: 10.1080/09205063.2021.1910920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nanotechnology has many potential applications in cancer treatment. For example, nano-drug delivery systems (NDDS) with high bioavailability, biodegradability, and biocompatibility have been developed, in order to increase the therapeutic effects of anticancer drugs. Among these NDDS, high-performance hydroxyapatite (HA) nanoparticles are rapidly advancing in the targeted cancer treatment due to their numerous benefits. Curcumin is an herbal metabolite that acts as a chemical inhibitor through the inhibition of tumor cells and the progression of many cancers. However, the poor bioavailability of curcumin is the most important challenge in using this substance. In this study, HA nanoparticles coated by chitosan were used as a pH-sensitive biopolymer to improve the efficiency and bioavailability of curcumin. For this purpose, HA nanoparticles were first synthesized by the sol-gel method. Then, a layer of chitosan was coated on it, and the curcumin drug was encapsulated in the nanocarrier, under controlled conditions. Techniques such as scanning electron microscopy (SEM), X-ray diffraction (XRD), and Fourier transform infrared spectroscopy (FTIR) were used to characterize the nanocarriers. In the second part, nano-drugs prepared by various bioassays were examined. For this purpose, the rate of cytotoxicity by the methyl-thiazol-tetrazolium (MTT) assay and the rate of apoptosis induction by the acridine orange and ethidium bromide (AO/EB) staining method on the brain carcinoma U87MG cell line were investigated.
Collapse
Affiliation(s)
- Katayon Hemmati
- Hormoz Research Center, University of Hormozgan, Bandar Abbas, Iran.,Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | | | - Saeed Hesaraki
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| | - Nader Nezafati
- Nanotechnology and Advanced Materials Department, Materials and Energy Research Center, Karaj, Iran
| |
Collapse
|