1
|
Zhang W, Qiao H, Cui J, Zhang D, Li Y. The redox homeostasis-reshapable hyaluronic acid-drug conjugate augments chemo-photodynamic therapy. Int J Biol Macromol 2025; 310:143465. [PMID: 40280526 DOI: 10.1016/j.ijbiomac.2025.143465] [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: 12/25/2024] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The tumor microenvironment poses significant challenges to reactive oxygen species treatment, and fails to retain small molecule drugs for extended periods, leading to low treatment efficacy. Here, a hyaluronic acid-based nanoplatform (HCCD) for combined chemotherapy and photodynamic therapy (PDT) is developed. The nanoplatform links chlorin e6 (Ce6) and doxorubicin (DOX) with hyaluronic acid through a glutathione (GSH)-sensitive disulfide bond, resulting their fluorescence quenching. Notably, their fluorescence and cytotoxicity are specifically activated in tumor cells, not normal ones, due to the endogenous GSH-mediated cleavage of disulfide bonds. Furthermore, PDT is activated in tumor cells with 660 nm laser, and GSH consumption reshapes redox homeostasis balance, thereby synergistically enhancing the anti-tumor efficacy with chemotherapy. HCCD predominantly accumulates in tumors in vivo, enabling precise localization and guidance for PDT and chemotherapy. This approach results in a significant reduction in tumor size, by a factor of 11 compared to the control group, with tumors nearly disappearing. Additionally, HCCD with the toll-like receptor 7 agonist imiquimod (R837) and anti-programmed death ligand 1 (anti-PD-L1) activated systemic immune response, suppressing distant tumor growth. Therefore, it's a promising strategy for precise tumor targeting and combination therapy, with potential in immunotherapy to inhibit primary and metastatic tumor growth.
Collapse
Affiliation(s)
- Wenxia Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, PR China
| | - Hui Qiao
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, PR China
| | - Jicheng Cui
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, PR China
| | | | - Yingqi Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, PR China; Department of Chemistry, College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China.
| |
Collapse
|
2
|
Dwivedi R, Bala R, Singh S, Sindhu RK. Catechins in cancer therapy: integrating traditional and complementary approaches. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2024-0472. [PMID: 39976450 DOI: 10.1515/jcim-2024-0472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Catechin is a group of bioactive flavonoids found in various plant sources such as tea, cocoa, and fruits. Recent studies have suggested that catechins has significant potential in preventing and treating cancer. Catechin exhibits a variety of biological activities that may contribute to its anticancer effects, including antioxidant, anti-inflammatory, and pro-apoptotic properties. Studies have demonstrated that catechin can inhibit cancer cell proliferation, induce cell cycle arrest, and promote apoptosis across multiple cancer types, including skin, breast, lung, liver, prostate, and colon cancers. Furthermore, catechin has shown the ability to inhibit angiogenesis, a critical process for tumor growth and metastasis, by restricting new blood vessel formation. Catechin's impact on cancer extends beyond its direct effects on cancer cells. It modulates various signaling pathways involved in cancer progression, such as those associated with cell survival, inflammation, and metastasis. Despite these promising findings, additional research is needed to clarify the precise mechanisms of catechin's anticancer action, optimal dosing strategies, and long-term safety in cancer prevention and treatment. This review will explore the current research landscape on tea polyphenols, particularly catechin, and their potential role in cancer prevention and therapy.
Collapse
Affiliation(s)
- Renu Dwivedi
- School of Pharmaceutical Sciences, Bahra University, Solan, Himachal Pradesh, India
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rajni Bala
- University School of Pharmaceutical Sciences, Rayat Bahra University, Mohali, Punjab, India
| | - Sumitra Singh
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| | - Rakesh K Sindhu
- Sharda School of Pharmacy, 193167 Sharda University , Greater Noida, UP, India
| |
Collapse
|
3
|
Yang M, Liu D, Tan Y, Chen J, Yang F, Mei C, Zeng Q, Lin Y, Li D. Polyoxometalate-based injectable coacervate inhibits HCC metastasis after incomplete radiofrequency ablation via scavenging ROS. J Nanobiotechnology 2025; 23:47. [PMID: 39871237 PMCID: PMC11773879 DOI: 10.1186/s12951-024-02989-1] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Incomplete radiofrequency ablation (iRFA) stimulates residual hepatocellular carcinoma (HCC) metastasis, leading to a poor prognosis for patients. Therefore, it is imperative to develop an effective therapeutic strategy to prevent iRFA-induced HCC metastasis. RESULTS Our study revealed that iRFA induced an abnormal increase in ROS levels within residual HCC, which enhanced tumor cell invasiveness and promoted macrophage M2 polarization, ultimately facilitating HCC metastasis. Molybdenum-based polyoxometalate (POM) is an excellent ROS-scavenging nanocluster, but its size is too small to be easily cleared by the kidneys, limiting its effectiveness in scavenging iRFA-induced ROS. To overcome this limitation, we synthesized an injectable POM-loaded coacervate delivery system named POM@Coa, which can sustainably scavenge iRFA-induced ROS by slowly releasing POM. POM@Coa markedly reduced HCC invasiveness, reversed macrophage polarization from M2 to M1, and promoted the infiltration and activation of CD8+ T cells, ultimately inhibiting HCC metastasis. Importantly, POM@Coa showed superior therapeutic efficacy to free POM in the absence of systemic toxicity. CONCLUSIONS POM@Coa exhibits the potential to decrease HCC invasiveness and activate anti-tumor immunity, opening up new avenues for the safe and effective treatment and prevention of HCC metastasis when combined with RFA.
Collapse
Affiliation(s)
- Meilin Yang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China
| | - Die Liu
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China
| | - Yan Tan
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China
| | - Jieting Chen
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China
| | - Fan Yang
- Department of Pediatrics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
| | - Chaoming Mei
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China
| | - Qi Zeng
- Cancer Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China.
| | - Yong Lin
- Department of Psychiatry, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China.
| | - Dan Li
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China.
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, China.
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, 519000, China.
| |
Collapse
|
4
|
Jiao C, Qiu J, Gong C, Li X, Liang H, He C, Cen S, Xie Y. Ganoderma lucidum extract reverses multidrug resistance in breast cancer cells through inhibiting ATPase activity of the P-glycoprotein via MAPK/ERK signaling pathway. Exp Cell Res 2025; 444:114355. [PMID: 39613022 DOI: 10.1016/j.yexcr.2024.114355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024]
Abstract
Breast cancer represents a persistent global health challenge, with multidrug resistance (MDR) posing a significant obstacle to effective treatment. In this study, we investigate the potential of Ganoderma lucidum extract (GLE) in reversing MDR in breast cancer and delve into the underlying mechanisms. We establish a robust in vitro 3D model of breast cancer with acquired MDR induced by paclitaxel. Utilizing the CCK-8 method, we assess the impact of GLE on cytotoxic drug sensitivity to determine its in vitro MDR reversal activity. Our results reveal that GLE enhances the toxicity of paclitaxel in breast cancer cells by inhibiting the ATPase activity of P-glycoprotein (P-gp) and increasing the intracellular and extracellular excretion of P-gp substrates, all without significantly altering P-gp protein expression. Additionally, GLE inhibits the phosphorylation of ERK1/2, suggesting that the enhanced sensitivity of breast cancer cells to paclitaxel by GLE is associated with the MAPK pathway. These findings indicate that GLE may inhibit P-gp-mediated drug efflux via the MAPK pathway, thus effectively overcoming paclitaxel resistance in breast cancer. This study provides valuable insights into the potential clinical applications of GLE in reversing multidrug resistance, offering hope for improved breast cancer treatment strategies.
Collapse
Affiliation(s)
- Chunwei Jiao
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China
| | - Jinshou Qiu
- Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, Fujian, 363000, PR China
| | - Congcong Gong
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; South China University of Technology, PR China
| | - Xiaoyi Li
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Huijia Liang
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Chunyan He
- Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China
| | - Sien Cen
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China
| | - Yizhen Xie
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, PR China; Guangdong Yuewei Bioscience Co., Ltd., Zhaoqing, 526000, PR China.
| |
Collapse
|
5
|
Jeeva P, Muthusamy A, Kesavan swaminathan J. Deciphering Structural Dynamics of Atherosclerosis Proteins: Insights from Crataegus oxyacantha Phytochemicals that Interceded Functional and Structural Changes in Targeted Atherosclerotic Proteins. ACS OMEGA 2024; 9:48159-48172. [PMID: 39676950 PMCID: PMC11635474 DOI: 10.1021/acsomega.4c04975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024]
Abstract
Atherosclerosis (ASC) is characterized by foam cell-mediated plaque formation, vascular endothelial inflammation, and lipidosis and is the rudimentary cause of cardiovascular diseases. This is the pre-eminent global factor of mortality. This etiological paradigm is significantly influenced by several proteins, where 23 pivotal proteins involved in ASC were meticulously gleaned on the basis of literature studies. The crux of the present study was aimed to probe the drugability of four active phytochemicals from Crataegus oxyacantha (COC): epicatechin, gallate, tyramine, and vitexin against the selected 23 proteins. The molecular docking analysis was judiciously administered via Glide, the binding free energy was calculated in detail utilizing the prime molecular mechanics-generalized Born surface area (MM-GBSA) module, and a deeper comprehensive investigation of protein-ligand dynamic associations was elucidated through Desmond. Drawing from the upper echelons of our docking results, the molecular dynamics simulation outcomes revealed that the macrophage migration inhibitory factor and prethrombin-1 showed persistent binding nature with gallate. The bioactive compound known as gallate sourced from COC shows the best molecular association with pivotal proteins involved in ASC and has a promising therapeutic potential for drug development endeavors.
Collapse
Affiliation(s)
- Praveen Jeeva
- Department
of Bioinformatics, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | - Anusuyadevi Muthusamy
- Department
of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamilnadu 620024, India
| | | |
Collapse
|
6
|
Sayed Tabatabaei M, Sayed Tabatabaei FA, Moghimi HR. Drug self-delivery systems: A comprehensive review on small molecule nanodrugs. BIOIMPACTS : BI 2024; 15:30161. [PMID: 40161942 PMCID: PMC11954755 DOI: 10.34172/bi.30161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 04/02/2025]
Abstract
Drug self-delivery systems are nanostructures composed of a drug as the main structural unit, having the ability of intracellular trafficking with no additional carrier. In these systems, the drug itself undertakes the functional and structural roles; thereby, the ancillary role of excipients and carrier-related limitations are circumvented and therapeutic effect is achieved at a much lower dose. Such advantages -which are mainly but not exclusively beneficial in cancer treatment- have recently led to an upsurge of research on these systems. Subsequently, various terminologies were utilized to describe them, referring to the same concept with different words. However, not all the systems developed based on the self-delivery approach are introduced using one of these keywords. Using a scoping strategy, this review aims to encompass the systems that have been developed as yet -inspired by the concept of self-delivery- and classify them in a coherent taxonomy. Two main groups are introduced based on the type of building blocks: small molecule-based nanomedicines and self-assembling hybrid prodrugs. Due to the diversity, covering the whole gamut of topics is beyond the scope of a single article, and, inevitably, the latter is just briefly introduced here, whereas the features of the former group are meticulously presented. Depending on whether the drug is merely a carrier for itself or carries a second drug as cargo, two classes of small molecule-based nanomedicines are defined (i.e., pure nanodrugs and carrier-mimicking systems, respectively), each having sub-branches. After introducing each branch and giving some examples, possible strategies for designing each particular system are visually displayed. The resultant mind map can create a macro view of the taken path and its prospects, give a profound insight into opportunities, spark new ideas, and facilitate overcoming obstacles. Taken together, one can foresee a brilliant future for self-delivery systems as a pioneering candidate for the next generation of drug delivery systems.
Collapse
Affiliation(s)
- Mahsa Sayed Tabatabaei
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamid Reza Moghimi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Zheng Y, Chen X, Wang Y, Chen Z, Wu D. Phenolic-enabled nanotechnology: a new strategy for central nervous system disease therapy. J Zhejiang Univ Sci B 2024; 25:890-913. [PMID: 39420524 PMCID: PMC11494163 DOI: 10.1631/jzus.b2300839] [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: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 10/19/2024]
Abstract
Polyphenolic compounds have received tremendous attention in biomedicine because of their good biocompatibility and unique physicochemical properties. In recent years, phenolic-enabled nanotechnology (PEN) has become a hotspot of research in the medical field, and many promising studies have been reported, especially in the application of central nervous system (CNS) diseases. Polyphenolic compounds have superior anti-inflammatory and antioxidant properties, and can easily cross the blood‒brain barrier, as well as protect the nervous system from metabolic damage and promote learning and cognitive functions. However, although great advances have been made in this field, a comprehensive review regarding PEN-based nanomaterials for CNS therapy is lacking. A systematic summary of the basic mechanisms and synthetic strategies of PEN-based nanomaterials is beneficial for meeting the demand for the further development of novel treatments for CNS diseases. This review systematically introduces the fundamental physicochemical properties of PEN-based nanomaterials and their applications in the treatment of CNS diseases. We first describe the different ways in which polyphenols interact with other substances to form high-quality products with controlled sizes, shapes, compositions, and surface chemistry and functions. The application of PEN-based nanomaterials in the treatment of CNS diseases is then described, which provides a reference for subsequent research on the treatment of CNS diseases.
Collapse
Affiliation(s)
- Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
- Zhejiang Rehabilitation Medical Center, the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310009, China. ,
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. ,
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
8
|
Setia A, Challa RR, Vallamkonda B, Satti P, Mehata AK, Priya V, Kumar S, Muthu MS. Nanomedicine And Nanotheranostics: Special Focus on Imaging of Anticancer Drugs Induced Cardiac Toxicity. Nanotheranostics 2024; 8:473-496. [PMID: 38961885 PMCID: PMC11217786 DOI: 10.7150/ntno.96846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 04/17/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiotoxicity, the often-overlooked second leading cause of death in cancer patients, has been associated with certain anticancer drugs. These drugs can induce cardiac damage through various pathways, and their adverse effects on the heart are not fully understood. Cardiotoxicity is a major issue in cancer treatment, particularly with chemotherapeutics, because it can cause cardiac dysfunction such as hypotension, heart failure, and even death. Doxorubicin, 5-fluorouracil, and trastuzumab, all of which are very potent anticancer drugs, are known to cause cardiotoxicity. When it comes to lowering cardiotoxicity and alleviating the harmful effects of chemotherapy medications, nanomedicine has the potential to transport therapeutic molecules. Nanotheranostics offers novel options for identifying and treating cardiotoxicity resulting from a wide range of substances, including anticancer medications. Additionally, theranostics platforms such as micellar systems, carbon-based nanomedicine, solid lipid nanoparticles, polymeric nanoparticles, and liposomes can transport chemotherapeutic medications while minimising their cardiotoxicity. The present level of understanding of the molecular and cellular processes that lead to cardiotoxicity in reaction to both traditional chemotherapy and targeted drug delivery systems is summarised in this article. This review delves into nanomedicine and nanotheranostics, with an emphasis on reducing anticancer medication-induced cardiac toxicity. Nanotheranostics provide potential solutions for early diagnosis and tailored therapy of heart injury by combining diagnostic and therapeutic capabilities into nanomedicine.
Collapse
Affiliation(s)
- Aseem Setia
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Randheer Reddy Challa
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN's Foundation for Science, Technology & Research, Vadlamudi-522213, Andhra Pradesh, India
| | - Bhaskar Vallamkonda
- Department of Pharmaceutical Science, School of Applied Sciences and Humanities, VIGNAN's Foundation for Science, Technology & Research, Vadlamudi-522213, Andhra Pradesh, India
| | - Phanikumarreddy Satti
- Department of Chemistry, Acharya Nagarjuna University, Guntur- 522510, Andhra Pradesh, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Vishnu Priya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| | - Senthil Kumar
- Pharmacy Services, Homi Bhabha Cancer Hospital & Mahamana Pandit Madan Mohan Malaviya Cancer Centre, Varanasi-221005, India
| | - Madaswamy S. Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi-221005, India
| |
Collapse
|
9
|
Yao SY, Ying AK, Jiang ZT, Cheng YQ, Geng WC, Hu XY, Cai K, Guo DS. Single Molecular Nanomedicines Based on Macrocyclic Carrier-Drug Conjugates for Concentration-Independent Encapsulation and Precise Activation of Drugs. J Am Chem Soc 2024; 146:14203-14212. [PMID: 38733560 DOI: 10.1021/jacs.4c03238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Nanomedicines often rely on noncovalent self-assembly and encapsulation for drug loading and delivery. However, challenges such as reproducibility issues due to the multicomponent nature, off-target activation caused by premature drug release, and complex pharmacokinetics arising from assembly dissociation have hindered their clinical translation. In this study, we introduce an innovative design concept termed single molecular nanomedicine (SMNM) based on macrocyclic carrier-drug conjugates. Through the covalent linkage of two chemotherapy drugs to a hypoxia-cleavable macrocyclic carrier, azocalix[4]arene, we obtained two self-included complexes to serve as SMNMs. The intramolecular inclusion feature of the SMNMs has not only demonstrated comprehensive shielding and protection for the drugs but also effectively prevented off-target drug leakage, thereby significantly reducing their side effects and enhancing their antitumor therapeutic efficacy. Additionally, the attributes of being a single component and molecularly dispersed confer advantages such as ease of preparation and good reproducibility for SMNMs, which is desirable for clinical applications.
Collapse
Affiliation(s)
- Shun-Yu Yao
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - An-Kang Ying
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yuan-Qiu Cheng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Kang Cai
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi 844000, China
| |
Collapse
|
10
|
Peng X, McClements DJ, Liu X, Liu F. EGCG-based nanoparticles: synthesis, properties, and applications. Crit Rev Food Sci Nutr 2024; 65:2177-2198. [PMID: 38520117 DOI: 10.1080/10408398.2024.2328184] [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] [Indexed: 03/25/2024]
Abstract
(-)-Epigallocatechin-3-gallate (EGCG) is a natural phenolic substance found in foods and beverages (especially tea) that exhibits a broad spectrum of biological activities, including antioxidant, antimicrobial, anti-obesity, anti-inflammatory, and anti-cancer properties. Its potential in cardiovascular and brain health has garnered significant attention. However, its clinical application remains limited due to its poor physicochemical stability and low oral bioavailability. Nanotechnology can be used to improve the stability, efficacy, and pharmacokinetic profile of EGCG by encapsulating it within nanoparticles. This article reviews the interactions of EGCG with various compounds, the synthesis of EGCG-based nanoparticles, the functional attributes of these nanoparticles, and their prospective applications in drug delivery, diagnosis, and therapy. The potential application of nanoencapsulated EGCG in functional foods and beverages is also emphasized. Top-down and bottom-up approaches can be used to construct EGCG-based nanoparticles. EGCG-based nanoparticles exhibit enhanced stability and bioavailability compared to free EGCG, making them promising candidates for biomedical and food applications. Notably, the non-covalent and covalent interactions of EGCG with other substances significantly contribute to the improved properties of these nanoparticles. EGCG-based nanoparticles appear to have a wide range of applications in different industries, but further research is required to enhance their efficacy and ensure their safety.
Collapse
Affiliation(s)
- Xiaoke Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | | | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
11
|
Bakun P, Mlynarczyk DT, Koczorowski T, Cerbin-Koczorowska M, Piwowarczyk L, Kolasiński E, Stawny M, Kuźmińska J, Jelińska A, Goslinski T. Tea-break with epigallocatechin gallate derivatives - Powerful polyphenols of great potential for medicine. Eur J Med Chem 2023; 261:115820. [PMID: 37776575 DOI: 10.1016/j.ejmech.2023.115820] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 10/02/2023]
Abstract
Epigallocatechin gallate (EGCG) is a polyphenol present in green tea (Camellia sinensis), which has revealed anti-cancer effects toward a variety of cancer cells in vitro and protective potential against neurodegenerative diseases such as Alzheimer's and Parkinson's. Unfortunately, EGCG presents disappointing bioavailability after oral administration, primarily due to its chemical instability and poor absorption. Due to these limitations, EGCG is currently not used in medication, but only as a dietary supplement in the form of green tea extract. Therefore, it needs further modifications before being considered suitable for extensive medical applications. In this article, we review the scientific literature about EGCG derivatives focusing on their biological properties and potential medical applications. The most common chemical modifications of epigallocatechin gallate rely on introducing fatty acid chains or sugar molecules to its chemical structure to modify solubility. Another frequently employed procedure is based on blocking EGCG's hydroxyl groups with various substituents. Novel derivatives reveal interesting properties, of which, antioxidant, anti-inflammatory, antitumor and antimicrobial, are especially important. It is worth noting that the most promising EGCG derivatives present higher stability and activity than base EGCG.
Collapse
Affiliation(s)
- Paweł Bakun
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland.
| | - Dariusz T Mlynarczyk
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Tomasz Koczorowski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Magdalena Cerbin-Koczorowska
- Chair and Department of Medical Education, Poznan University of Medical Sciences, Rokietnicka 7, Poznań, 60-806, Poland; Edinburgh Medical School: Medical Education, University of Edinburgh, Chancellor's Building, EH16 4SB, Edinburgh, Scotland, United Kingdom
| | - Ludwika Piwowarczyk
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Emil Kolasiński
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Maciej Stawny
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Joanna Kuźmińska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Anna Jelińska
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6, Poznań, 60-780, Poland.
| |
Collapse
|
12
|
Zhang T, Li N, Wang R, Sun Y, He X, Lu X, Chu L, Sun K. Enhanced therapeutic efficacy of doxorubicin against multidrug-resistant breast cancer with reduced cardiotoxicity. Drug Deliv 2023; 30:2189118. [PMID: 36919676 PMCID: PMC10026743 DOI: 10.1080/10717544.2023.2189118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX), a commonly used anti-cancer drug, is limited by its cardiotoxicity and multidrug resistance (MDR) of tumor cells. Epigallocatechin gallate (EGCG), a natural antioxidant component, can effectively reduce the cardiotoxicity of DOX. Meanwhile, EGCG can inhibit the expression of P-glycoprotein (P-gp) and reverse the MDR of tumor cells. In this study, DOX is connected with low molecular weight polyethyleneimine (PEI) via hydrazone bond to get the pH-sensitive PEI-DOX, which is then combined with EGCG to prevent the cardiotoxicity of DOX and reverse the MDR of cancer cells. In addition, folic acid (FA) modified polyethylene glycol (PEG) (PEG-FA) is added to get the targeted system PEI-DOX/EGCG/FA. The MDR reversal and targeting ability of PEI-DOX/EGCG/FA is performed by cytotoxicity and in vivo anti-tumor activity on multidrug resistant MCF-7 cells (MCF-7/ADR). Additionally, we investigate the anti-drug resistant mechanism by Western Blot. The ability of EGCG to reduce DOX cardiotoxicity is confirmed by cardiotoxicity assay. In conclusion, PEI-DOX/EGCG/FA can inhibit the expression of P-gp and reverse the MDR in tumor cells. It also shows the ability of remove oxygen free radicals effectively to prevent the cardiotoxicity of DOX.
Collapse
Affiliation(s)
- Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Xiaoyan He
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| |
Collapse
|
13
|
Luo X. Nanobiotechnology-based strategies in alleviation of chemotherapy-mediated cardiotoxicity. ENVIRONMENTAL RESEARCH 2023; 238:116989. [PMID: 37633635 DOI: 10.1016/j.envres.2023.116989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
The cardiovascular diseases have been among the most common malignancies and the first leading cause of death, even higher than cancer. The cardiovascular diseases can be developed as a result of cardiac dysfunction and damages to heart tissue. Exposure to toxic agents and chemicals that induce cardiac dysfunction has been of interest in recent years. The chemotherapy drugs are commonly used for cancer therapy and in these patients, cardiovascular diseases have been widely observed that is due to negative impact of chemotherapy drugs on the heart. These drugs increase oxidative damage and inflammation, and mediate apoptosis and cardiac dysfunction. Hence, nanotechnological approaches have been emerged as new strategies in attenuation of chemotherapy-mediated cardiotoxicity. The first advantage of nanoparticles can be explored in targeted and selective delivery of drugs to reduce their accumulation in heart tissue. Nanostructures can deliver bioactive and therapeutic compounds in reducing cardiotoxicity and alleviation toxic impacts of chemotherapy drugs. The functionalization of nanostructures increases their selectivity against tumor cells and reduces accumulation of drugs in heart tissue. The bioplatforms such as chitosan and alginate nanostructures can also deliver chemotherapy drugs and reduce their cardiotoxicity. The function of nanostructures is versatile in reduction of cardiotoxicity by chemotherapy drugs and new kind of platforms is hydrogels that can mediate sustained release of drug to reduce its toxic impacts on heart tissue. The various kinds of nanoplatforms have been developed for alleviation of cardiotoxicity and their future clinical application depends on their biocompatibility. High concentration level of chitosan nanoparticles can stimulate cardiotoxicity. Therefore, if nanotechnology is going to be deployed for drug delivery and reducing cardiotoxicity, the first pre-requirement is to lack toxicity on normal cells and have high biocompatibility.
Collapse
Affiliation(s)
- Xuanming Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China.
| |
Collapse
|
14
|
Yu M, Hu S, Tang B, Yang H, Sun D. Engineering Escherichia coli Nissle 1917 as a microbial chassis for therapeutic and industrial applications. Biotechnol Adv 2023; 67:108202. [PMID: 37343690 DOI: 10.1016/j.biotechadv.2023.108202] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/19/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Genetically engineered microbes, especially Escherichia coli, have been widely used in the biosynthesis of proteins and metabolites for medical and industrial applications. As a traditional probiotic with a well-established safety record, E. coli Nissle 1917 (EcN) has recently emerged as a microbial chassis for generating living therapeutics, drug delivery vehicles, and microbial platforms for industrial production. Despite the availability of genetic tools for engineering laboratory E. coli K-12 and B strains, new genetic engineering systems are still greatly needed to expand the application range of EcN. In this review, we have summarized the latest progress in the development of genetic engineering systems in EcN, as well as their applications in the biosynthesis and delivery of valuable small molecules and biomacromolecules of medical and/or industrial interest, followed by a glimpse of how this rapidly growing field will evolve in the future.
Collapse
Affiliation(s)
- Mingjing Yu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Shilong Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Biao Tang
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Hua Yang
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Dongchang Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
15
|
Wang X, Li J, Chen R, Li T, Chen M. Active Ingredients from Chinese Medicine for Combination Cancer Therapy. Int J Biol Sci 2023; 19:3499-3525. [PMID: 37497002 PMCID: PMC10367560 DOI: 10.7150/ijbs.77720] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/26/2023] [Indexed: 07/28/2023] Open
Abstract
Combination therapy against cancer has gained increasing attention because it can help to target multiple pathways to tackle oncologic progression and improve the limited antitumor effect of single-agent therapy. Chinese medicine has been studied extensively in cancer therapy and proven to be efficacious in many cases due to its wide spectrum of anticancer activities. In this review, we aim to summarize the recent progress of active ingredients from Chinese medicine (AIFCM) in combination with various cancer therapeutic modalities, including chemotherapy, gene therapy, radiotherapy, phototherapy and immunotherapy. In addition to highlighting the potential contribution of AIFCM in combination cancer therapy, we also elucidate the underlying mechanisms behind their synergistic effect and improved anticancer efficacy, thereby encouraging the inclusion of these AIFCM as part of effective armamentarium in fighting intractable cancers. Finally, we present the challenges and future perspectives of AIFCM combination therapy as a feasible and promising strategy for the optimization of cancer treatment and better clinical outcomes.
Collapse
Affiliation(s)
- Xuan Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Jing Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Ruie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, 999078, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau, 999078, China
| |
Collapse
|
16
|
Feng M, Dai X, Yang C, Zhang Y, Tian Y, Qu Q, Sheng M, Li Z, Peng X, Cen S, Shi X. Unification of medicines and excipients: The roles of natural excipients for promoting drug delivery. Expert Opin Drug Deliv 2023; 20:597-620. [PMID: 37150753 DOI: 10.1080/17425247.2023.2210835] [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: 02/06/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Drug delivery systems (DDSs) formed by natural active compounds be instrumental in developing new green excipients and novel DDS from natural active compounds (NACs). 'Unification of medicines and excipients'(UME), the special inherent nature of the natural active compounds, provides the inspiration and conduction to achieve this goal. AREAS COVERED This review summarizes the typical types of NACs from herbal medicine, such as saponins, flavonoids, polysaccharides, etc. that act as excipients and their main application in DDS. The comparison of the drug delivery systems formed by NACs and common materials and the primary formation mechanisms of these NACs are also introduced to provide a deepened understanding of their performance in DDS. EXPERT OPINION Many natural bioactive compounds, such as saponins, polysaccharides, etc. have been used in DDS. Diversity of structure and pharmacological effects of NACs turn out the unique advantages in improving the performance of DDSs like targeting ability, adhesion, encapsulation efficiency(EE), etc. and enhancing the bioavailability of loaded drugs.
Collapse
Affiliation(s)
- Minfang Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxing Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing, China
| | - Cuiting Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Qingsong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Mengke Sheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixun Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinhui Peng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shuai Cen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing, China
| |
Collapse
|
17
|
Chen W, Liu M, Yang H, Nezamzadeh-Ejhieh A, Lu C, Pan Y, Liu J, Bai Z. Recent Advances of Fe(III)/Fe(II)-MPNs in Biomedical Applications. Pharmaceutics 2023; 15:pharmaceutics15051323. [PMID: 37242566 DOI: 10.3390/pharmaceutics15051323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Metal-phenolic networks (MPNs) are a new type of nanomaterial self-assembled by metal ions and polyphenols that have been developed rapidly in recent decades. They have been widely investigated, in the biomedical field, for their environmental friendliness, high quality, good bio-adhesiveness, and bio-compatibility, playing a crucial role in tumor treatment. As the most common subclass of the MPNs family, Fe-based MPNs are most frequently used in chemodynamic therapy (CDT) and phototherapy (PTT), where they are often used as nanocoatings to encapsulate drugs, as well as good Fenton reagents and photosensitizers to improve tumor therapeutic efficiency substantially. In this review, strategies for preparing various types of Fe-based MPNs are first summarized. We highlight the advantages of Fe-based MPNs under the different species of polyphenol ligands for their application in tumor treatments. Finally, some current problems and challenges of Fe-based MPNs, along with a future perspective on biomedical applications, are discussed.
Collapse
Affiliation(s)
- Weipeng Chen
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523700, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Miao Liu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Hanping Yang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | | | - Chengyu Lu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Ying Pan
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523700, China
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Jianqiang Liu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
- Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Zhi Bai
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523700, China
| |
Collapse
|
18
|
Wang L, Li P, Feng K. EGCG adjuvant chemotherapy: Current status and future perspectives. Eur J Med Chem 2023; 250:115197. [PMID: 36780831 DOI: 10.1016/j.ejmech.2023.115197] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
The resistance of cancer cells to chemotherapeutic drugs greatly reduces the therapeutic effect in cancer patients, and the toxic side effects caused by chemotherapy also seriously affect the quality of life of patients. The combination of epigallocatechin-3-gallate (EGCG), the main active ingredient in tea, with cisplatin, 5-FU, doxorubicin and paclitaxel enhances their sensitizing effect on tumors and combats the drug resistance of cancer cells. These effects seem to be mediated by a variety of mechanisms, including combating drug resistance mediated by cancer stem cells, enhancing drug sensitivity, inducing cell cycle arrest and apoptosis, and blocking angiogenesis. In addition, EGCG can suppress a series of adverse effects caused by chemotherapy, such as gastrointestinal disorders, nephrotoxicity and cardiotoxicity, through its anti-inflammatory and antioxidant effects and improve the quality of life of patients. However, the low bioavailability and off-target effects of EGCG and its reactivity with some chemotherapeutic agents limit its clinical application. The nanomodification of EGCG and chemotherapeutic drugs not only enhances the antitumor activity but also prolongs the survival time of tumor-bearing mice, and has the advantage of low toxicity. Therefore, this review aims to discuss the current status and challenges regarding the use of EGCG in combination with chemotherapy drugs in the treatment of cancer. In general, EGCG is a promising adjuvant for chemotherapy.
Collapse
Affiliation(s)
- Lin Wang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, 518118, Guangdong, China
| | - Penghui Li
- Center for Health Research, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, Guangdong, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kun Feng
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen, 518118, Guangdong, China.
| |
Collapse
|
19
|
Andreazzoli F, Bonucci M. Integrative Hematology: State of the Art. Int J Mol Sci 2023; 24:ijms24021732. [PMID: 36675247 PMCID: PMC9864076 DOI: 10.3390/ijms24021732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Blood cancers are a group of diseases with thus far frequently poor prognosis. Although many new drugs, including target therapies, have been developed in recent years, there is still a need to expand our therapeutic armamentarium to better deal with these diseases. Integrative hematology was conceived as a discipline that enriches the patient's therapeutic possibilities with the use of supplements, vitamins and a nutritional approach aiming at improving the response to therapies and the clinical outcome. We will analyze the substances that have proved most useful in preclinical and clinical studies in some of the most frequent blood diseases or in those where these studies are more numerous; the importance of the nutritional approach and the role of the intestinal microbiota will also be emphasized.
Collapse
Affiliation(s)
- Francesca Andreazzoli
- Department of Hematology, Versilia’s Hospital, Viale Aurelia, 335, 55049 Camaiore, Italy
- Correspondence:
| | - Massimo Bonucci
- Association for Research on Integrative Oncology Therapies (ARTOI), Via Ludovico Micara, 73, 00165 Rome, Italy
| |
Collapse
|
20
|
Bae KH, Lai F, Oruc B, Osato M, Chen Q, Kurisawa M. Self-Assembled Daunorubicin/Epigallocatechin Gallate Nanocomplex for Synergistic Reversal of Chemoresistance in Leukemia. Int J Mol Sci 2022; 24:ijms24010381. [PMID: 36613821 PMCID: PMC9820275 DOI: 10.3390/ijms24010381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Chemoresistance is one of the major challenges for the treatment of acute myeloid leukemia. Epigallocatechin gallate (EGCG), a bioactive polyphenol from green tea, has attracted immense interest as a potential chemosensitizer, but its application is limited due to the need for effective formulations capable of co-delivering EGCG and anti-leukemic drugs. Herein, we describe the formation and characterization of a micellar nanocomplex self-assembled from EGCG and daunorubicin, an anthracycline drug for the first-line treatment of acute myeloid leukemia. This nanocomplex was highly stable at pH 7.4 but stimulated to release the incorporated daunorubicin at pH 5.5, mimicking an acidic endosomal environment. More importantly, the nanocomplex exhibited superior cytotoxic efficacy against multidrug-resistant human leukemia cells over free daunorubicin by achieving a strong synergism, as supported by median-effect plot analysis. The observed chemosensitizing effect was in association with enhanced nucleus accumulation of daunorubicin, elevation of intracellular reactive oxygen species and caspase-mediated apoptosis induction. Our study presents a promising strategy for circumventing chemoresistance for more effective leukemia therapy.
Collapse
Affiliation(s)
- Ki Hyun Bae
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Fritz Lai
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore
| | - Betul Oruc
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Motomi Osato
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, The Proteos, Singapore 138673, Singapore
| | - Motoichi Kurisawa
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
- Graduate School of Advanced Science and Technology, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi 923-1292, Ishikawa, Japan
- Correspondence:
| |
Collapse
|
21
|
Li B, Shao H, Gao L, Li H, Sheng H, Zhu L. Nano-drug co-delivery system of natural active ingredients and chemotherapy drugs for cancer treatment: a review. Drug Deliv 2022; 29:2130-2161. [PMID: 35815678 PMCID: PMC9275501 DOI: 10.1080/10717544.2022.2094498] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy drugs have been used for a long time in the treatment of cancer, but serious side effects are caused by the inability of the drug to be solely delivered to the tumor when treating cancer with chemotherapy. Natural products have attracted more and more attention due to the antitumor effect in multiple ways, abundant resources and less side effects. Therefore, the combination of natural active ingredients and chemotherapy drugs may be an effective antitumor strategy, which can inhibit the growth of tumor and multidrug resistance, reduce side effects of chemotherapy drugs. Nano-drug co-delivery system (NDCDS) can play an important role in the combination of natural active ingredients and chemotherapy drugs. This review provides a comprehensive summary of the research status and application prospect of nano-delivery strategies for the combination of natural active ingredients and chemotherapy drugs, aiming to provide a basis for the development of anti-tumor drugs.
Collapse
Affiliation(s)
- Bingqian Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huili Shao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huan Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
22
|
Talib WH, Abuawad A, Thiab S, Alshweiat A, Mahmod AI. Flavonoid-based nanomedicines to target tumor microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Li T, Liu Z, Fu X, Chen Y, Zhu S, Zhang J. Co-delivery of Interleukin-12 and Doxorubicin Loaded Nano-delivery System for Enhanced Immunotherapy with Polarization toward M1-type Macrophages. Eur J Pharm Biopharm 2022; 177:175-183. [PMID: 35811038 DOI: 10.1016/j.ejpb.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/21/2022] [Accepted: 07/04/2022] [Indexed: 01/07/2023]
Abstract
Chemo-immunotherapy has gained increasing attention as one of the most promising combination therapy strategies to battle cancer. In this study, the therapeutic nanoparticles (TNPs) co-delivering doxorubicin (DOX) and IL-12 (IL-12) were developed for chemo-immunotherapy combination therapy on liver cancer. TNPs were synthesized based on the ionic interactions between cationic chitosan (Ch) and anionic poly-(glutamic acid) (PGA). DOX and IL-12 loaded in TNPs presented prolonged circulation in blood, efficient accumulation in tumors, and internalization in tumor cells. After that, DOX and IL-12 were co-released in the tumor microenvironment. The locally responsive property of TNPs could subsequently re-educate macrophages. More significantly, TNPs with no obvious side effects can remarkably inhibit the H22 tumor growth in vivo. A low dosage of loaded IL-12 in TNPs can effectively polarize macrophages toward the M1 phenotype to reduce tumor burden, further enhancing the antitumor efficacy. Our results suggest that the self-stabilized TNPs could be a secure and effective drug carrier for intravenous administration when deprived of protective agents.
Collapse
Affiliation(s)
- Tushuai Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214013, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214013, China; School of Food Science and Technology, Jiangnan University, Wuxi 214013, China
| | - Zhihong Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Sciences, Nanjing University, Nanjing 210033, China
| | - Xiao Fu
- Department of General Surgery, Institute of Translational Medicine, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214013, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214013, China; School of Food Science and Technology, Jiangnan University, Wuxi 214013, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214013, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214013, China.
| | - Jie Zhang
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou 215500, PR China.
| |
Collapse
|
24
|
Hybrid PEGylated chitosan/PLGA nanoparticles designed as pH-responsive vehicles to promote intracellular drug delivery and cancer chemotherapy. Int J Biol Macromol 2022; 210:565-578. [PMID: 35513093 DOI: 10.1016/j.ijbiomac.2022.04.209] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 12/18/2022]
Abstract
To achieve effective intracellular anticancer drug release for boosted antitumor efficacy, the acidity-responsive nanovehicles for doxorubicin (DOX) delivery were fabricated by tailor-made co-assembly of amphiphilic PEGylated chitosan20k and hydrophobic poly(lactic-co-glycolic acid) (PLGA) segments at pH 8.5. The attained DOX-loaded PEGylated chitosan20k/PLGA nanoparticles (DOX-PC20kPNs) were characterized to have a spherical shape composed of drug-encapsulated chitosan20k/PLGA-constituted solid core surrounded by hydrophilic PEG shells. Compared to non-pH-sensitive DOX-loaded PLGA nanoparticles (DOX-PNs), the DOX-PC20kPNs displayed outstanding colloidal stability under serum-containing condition and tended to swell in weak acidic milieu upon increased protonation of chitosan20k within hybrid cores, thus accelerating drug release. The in vitro cellular uptake and cytotoxicity studies revealed that the DOX-PC20kPNs after being endocytosed by prostate TRAMP-C1 cancer cells rapidly liberated drug, thus promoting drug accumulation in nuclei to enhance anticancer potency. Moreover, the hydrated PEG shells of DOX-PC20kPNs remarkably reduced their uptake by macrophage-like RAW264.7 cells. Importantly, in vivo animal findings showed that the DOX-PC20kPNs exhibited the capability of inhibiting TRAMP-C1 tumor growth superior to free hydrophobic DOX molecules and DOX-PNs, demonstrating the great potential in cancer chemotherapy.
Collapse
|
25
|
Guo W, Li Z, Huang H, Xu Z, Chen Z, Shen G, Li Z, Ren Y, Li G, Hu Y. VB12-Sericin-PBLG-IR780 Nanomicelles for Programming Cell Pyroptosis via Photothermal (PTT)/Photodynamic (PDT) Effect-Induced Mitochondrial DNA (mitoDNA) Oxidative Damage. ACS APPLIED MATERIALS & INTERFACES 2022; 14:17008-17021. [PMID: 35394753 DOI: 10.1021/acsami.1c22804] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Pyroptosis, a kind of programmed cell death involving inflammation, might be a powerful way to fight against tumors, for example, using immunotherapy. However, how to trigger pyroptosis in cancer cells is an important issue. Photothermal (PTT)/photodynamic (PDT) therapy is a crucial strategy for inducing cancer cell pyroptosis with noninvasiveness. In this work, a sericin derivative modified with poly(γ-benzyl-l-glutamate) (PBLG) could self-assemble and was stable in an aqueous environment. Furthermore, the sericin derivative was conjugated with the tumor-targeting agent VB12 and loaded with IR780. Finally, we successfully synthesized VB12-sericin-PBLG-IR780 nanomicelles. The as-designed nanomicelles showed appropriate particle sizes, spherical morphology, improved photothermal stability, and high photothermal conversion efficiency (∼40%), which generated reactive oxygen species (ROS) simultaneously. Through enhanced cellular uptake, VB12-sericin-PBLG-IR780 could deliver more IR780 into cancer cells. With near-infrared (NIR), the VB12-sericin-PBLG-IR780 could significantly inhibit the expression of ATP synthase, called ATP5MC3, followed by mitochondrial damage. The presence of mitochondrial reactive oxygen species (mitoROS) led to oxidative damage of mitochondrial DNA (mitoDNA), which further activates NLRP3/Caspase-1/gasdermin D (GSDMD)-dependent pyroptosis and could promote dendritic cell (DC) maturation by pyroptosis. Furthermore, our data showed that VB12-sericin-PBLG-IR780 could achieve a brilliant antitumor effect and could activate DC maturation, initiate T-cell recruiting, and prime adaptive antitumor efficiency. Overall, our well-prepared nanomicelles might offer a tumor-targeted approach for programmed cell pyroptosis and inducing antitumor immunity via photothermal PTT/PDT effect-induced mitoDNA oxidative damage.
Collapse
Affiliation(s)
- Weihong Guo
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhao Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Huilin Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhijun Xu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhian Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guodong Shen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenyuan Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingxin Ren
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanfeng Hu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
26
|
Alam Khan S, Jawaid Akhtar M. Structural modification and strategies for the enhanced doxorubicin drug delivery. Bioorg Chem 2022; 120:105599. [DOI: 10.1016/j.bioorg.2022.105599] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 12/29/2022]
|
27
|
Liu X, Rong P. Recent Advances of Manganese-Based Hybrid Nanomaterials for Cancer Precision Medicine. Front Oncol 2021; 11:707618. [PMID: 34722253 PMCID: PMC8548572 DOI: 10.3389/fonc.2021.707618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022] Open
Abstract
Cancer precision medicine (CPM) could tailor the best treatment for individual cancer patients, while imaging techniques play important roles in its application. With the characteristics of noninvasion, nonionized, radiation-free, multidimensional imaging function, and real-time monitoring, magnetic resonance imaging (MRI) is an effective way for early tumor detection, and it has become a tower of strength in CPM imaging techniques. Due to linkage with nephrogenic systemic fibrosis (NSF), gadolinium (Gd)-based contrast agent (CA), which was long used in MRI, has been restricted by the Food and Drug Administration (FDA). In this review, we would like to introduce the manganese (Mn)-based CAs that could significantly increase the safety of MRI CAs by realizing more superior performance and functions simultaneously in the diagnosis and treatment of tumors. Also, recent advances in Mn-based hybrid nanomaterials for CPM are summarized and discussed.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China.,Postdoctoral Research Station of Clinical Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengfei Rong
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Zheng M, Pan M, Zhang W, Lin H, Wu S, Lu C, Tang S, Liu D, Cai J. Poly(α-l-lysine)-based nanomaterials for versatile biomedical applications: Current advances and perspectives. Bioact Mater 2021; 6:1878-1909. [PMID: 33364529 PMCID: PMC7744653 DOI: 10.1016/j.bioactmat.2020.12.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/05/2023] Open
Abstract
Poly(α-l-lysine) (PLL) is a class of water-soluble, cationic biopolymer composed of α-l-lysine structural units. The previous decade witnessed tremendous progress in the synthesis and biomedical applications of PLL and its composites. PLL-based polymers and copolymers, till date, have been extensively explored in the contexts such as antibacterial agents, gene/drug/protein delivery systems, bio-sensing, bio-imaging, and tissue engineering. This review aims to summarize the recent advances in PLL-based nanomaterials in these biomedical fields over the last decade. The review first describes the synthesis of PLL and its derivatives, followed by the main text of their recent biomedical applications and translational studies. Finally, the challenges and perspectives of PLL-based nanomaterials in biomedical fields are addressed.
Collapse
Affiliation(s)
- Maochao Zheng
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Miao Pan
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Wancong Zhang
- The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxiabei Road, Shantou, 515041, China
| | - Huanchang Lin
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Shenlang Wu
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Shijie Tang
- The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxiabei Road, Shantou, 515041, China
| | - Daojun Liu
- Shantou University Medical College, 22 Xinling Road, Shantou, 515041, China
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, 33620, USA
| |
Collapse
|
29
|
Li Q, Dong Z, Chen M, Feng L. Phenolic molecules constructed nanomedicine for innovative cancer treatment. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
30
|
Shu J, Li X, Dang J, Liu Y, Duan S, Zhu R, Yin L, Chen Y. Drug resistance reversal by interventing cancer bioenergetics with spherical helical polypeptide-potented gene silencing. CHEMICAL ENGINEERING JOURNAL 2021; 414:128545. [DOI: 10.1016/j.cej.2021.128545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
31
|
Guo Y, Sun Q, Wu FG, Dai Y, Chen X. Polyphenol-Containing Nanoparticles: Synthesis, Properties, and Therapeutic Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007356. [PMID: 33876449 DOI: 10.1002/adma.202007356] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Polyphenols, the phenolic hydroxyl group-containing organic molecules, are widely found in natural plants and have shown beneficial effects on human health. Recently, polyphenol-containing nanoparticles have attracted extensive research attention due to their antioxidation property, anticancer activity, and universal adherent affinity, and thus have shown great promise in the preparation, stabilization, and modification of multifunctional nanoassemblies for bioimaging, therapeutic delivery, and other biomedical applications. Additionally, the metal-polyphenol networks, formed by the coordination interactions between polyphenols and metal ions, have been used to prepare an important class of polyphenol-containing nanoparticles for surface modification, bioimaging, drug delivery, and disease treatments. By focusing on the interactions between polyphenols and different materials (e.g., metal ions, inorganic materials, polymers, proteins, and nucleic acids), a comprehensive review on the synthesis and properties of the polyphenol-containing nanoparticles is provided. Moreover, the remarkable versatility of polyphenol-containing nanoparticles in different biomedical applications, including biodetection, multimodal bioimaging, protein and gene delivery, bone repair, antibiosis, and cancer theranostics is also demonstrated. Finally, the challenges faced by future research regarding the polyphenol-containing nanoparticles are discussed.
Collapse
Affiliation(s)
- Yuxin Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Qing Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Yunlu Dai
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, P. R. China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
32
|
Han JH, Kim M, Kim HJ, Jang SB, Bae SJ, Lee IK, Ryu D, Ha KT. Targeting Lactate Dehydrogenase A with Catechin Resensitizes SNU620/5FU Gastric Cancer Cells to 5-Fluorouracil. Int J Mol Sci 2021; 22:ijms22105406. [PMID: 34065602 PMCID: PMC8161398 DOI: 10.3390/ijms22105406] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Resistance to anticancer therapeutics occurs in virtually every type of cancer and becomes a major difficulty in cancer treatment. Although 5-fluorouracil (5FU) is the first-line choice of anticancer therapy for gastric cancer, its effectiveness is limited owing to drug resistance. Recently, altered cancer metabolism, including the Warburg effect, a preference for glycolysis rather than oxidative phosphorylation for energy production, has been accepted as a pivotal mechanism regulating resistance to chemotherapy. Thus, we investigated the detailed mechanism and possible usefulness of antiglycolytic agents in ameliorating 5FU resistance using established gastric cancer cell lines, SNU620 and SNU620/5FU. SNU620/5FU, a gastric cancer cell harboring resistance to 5FU, showed much higher lactate production and expression of glycolysis-related enzymes, such as lactate dehydrogenase A (LDHA), than those of the parent SNU620 cells. To limit glycolysis, we examined catechin and its derivatives, which are known anti-inflammatory and anticancer natural products because epigallocatechin gallate has been previously reported as a suppressor of LDHA expression. Catechin, the simplest compound among them, had the highest inhibitory effect on lactate production and LDHA activity. In addition, the combination of 5FU and catechin showed additional cytotoxicity and induced reactive oxygen species (ROS)-mediated apoptosis in SNU620/5FU cells. Thus, based on these results, we suggest catechin as a candidate for the development of a novel adjuvant drug that reduces chemoresistance to 5FU by restricting LDHA.
Collapse
Affiliation(s)
- Jung Ho Han
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea;
- Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan 50612, Korea;
| | - MinJeong Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea;
| | - Hyeon Jin Kim
- Department of Molecular Biology, College of Natural Science, Busan 46241, Korea; (H.J.K.); (S.B.J.)
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Science, Busan 46241, Korea; (H.J.K.); (S.B.J.)
| | - Sung-Jin Bae
- Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan 50612, Korea;
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine Kyungpook National University, Daegu 41566, Korea;
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea;
- Correspondence: (D.R.); (K.-T.H.)
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea;
- Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan 50612, Korea;
- Correspondence: (D.R.); (K.-T.H.)
| |
Collapse
|
33
|
Zong L, Wang H, Hou X, Fu L, Wang P, Xu H, Yu W, Dai Y, Qiao Y, Wang X, Yuan Q, Pang X, Han G, Pu X. A novel GSH-triggered polymeric nanomicelles for reversing MDR and enhancing antitumor efficiency of hydroxycamptothecin. Int J Pharm 2021; 600:120528. [PMID: 33781880 DOI: 10.1016/j.ijpharm.2021.120528] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/22/2021] [Accepted: 03/21/2021] [Indexed: 12/15/2022]
Abstract
Tumor multidrug resistance (MDR) is one of the main reasons for the failure of clinical chemotherapy. Here, a bio-responsive anti-drug-resistant polymer micelle that can respond to the reductive GSH in the tumor microenvironment (TME) for delivery of HCPT was designed. A new type of polymer with anti-drug resistance and anti-tumor effect was synthesized and used to encapsulated HCPT to form reduction-sensitive micelles (PDSAH) by a thin-film dispersion method. It is demonstrated that the micelle formulation improves the anti-tumor activity and biosafety of HCPT, and also plays a significant role in reversing the drug resistance, which contributes to inhibiting the tumor growth and prolonging the survival time of H22 tumor-bearing mice. The results indicate that this nanoplatform can serve as a flexible and powerful system for delivery of other drugs that are tolerated by tumors or bacteria.
Collapse
Affiliation(s)
- Lanlan Zong
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Haiyan Wang
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Xianqiao Hou
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Like Fu
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Peirong Wang
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Hongliang Xu
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Wenjie Yu
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Yuxin Dai
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Yonghui Qiao
- Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510632, China
| | - Qi Yuan
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Xiaobin Pang
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Guang Han
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Xiaohui Pu
- Institute of Pharmacy, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| |
Collapse
|
34
|
Fan R, Chen C, Hou H, Chuan D, Mu M, Liu Z, Liang R, Guo G, Xu J. Tumor Acidity and Near‐Infrared Light Responsive Dual Drug Delivery Polydopamine‐Based Nanoparticles for Chemo‐Photothermal Therapy. ADVANCED FUNCTIONAL MATERIALS 2021. [DOI: 10.1002/adfm.202009733] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Rangrang Fan
- Department of Neurosurgery West China Hospital Sichuan University Chengdu 610041 P. R. China
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Caili Chen
- School of Basic Medical Sciences Xinxiang Medical University Xinxiang Henan 453003 P. R. China
| | - Huan Hou
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Di Chuan
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Min Mu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Zhiyong Liu
- Department of Neurosurgery West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Ruichao Liang
- Department of Neurosurgery West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Gang Guo
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University and Collaborative Innovation Center for Biotherapy Chengdu 610041 P. R. China
| | - Jianguo Xu
- Department of Neurosurgery West China Hospital Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
35
|
Haghi A, Raissi H, Hashemzadeh H, Farzad F. Designing a high-performance smart drug delivery system for the synergetic co-absorption of DOX and EGCG on ZIF-8. RSC Adv 2020; 10:44533-44544. [PMID: 35517168 PMCID: PMC9058488 DOI: 10.1039/d0ra08123j] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/25/2020] [Indexed: 01/27/2023] Open
Abstract
Due to the extreme pore volume and valuable surface area, zeolitic imidazole frameworks (ZIFs) are promising vehicles that enhance the delivery of therapeutic agents to tissues. Furthermore, these nanoporous materials have high stability in the pH and temperature of the surrounding healthy cells (37 °C and pH = 7) and an exotic potential to deform in carcinogenic environment (T > 37 °C and pH ∼ 5.5), which make them perfect smart drug delivery vehicle candidates. In this work, a series of molecular dynamics (MD) and metadynamics simulations have been performed to gain molecular insight into the mechanisms involved in the process of co-loading of doxorubicin (DOX) and EpiGalloCatechin-3 Gallate (EGCG) on ZIF-8, which form a smart drug delivery system (SDDS). The obtained results revealed that DOX was adsorbed on the carrier mostly through electrostatic interactions (E coul = ∼-1200 kJ mol-1, E tot = -1700 kJ mol-1), and EGCG was stacked on ZIF-8 mainly via van der Waals interactions (E L-J = ∼-600 kJ mol-1, E tot = ∼-1200 kJ mol-1). It is worth mentioning that the drug-drug L-J interactions (E L-J = ∼500 kJ mol-1) were also important in the co-loading process. The insertion of DOX and EGCG as additive agents to the initial ZIF-8/EGCG and ZIF-8/DOX systems led to the enhancement of the drug-carrier pair interactions to about ∼-2300 kJ mol-1 and ∼-2000 kJ mol-1, respectively. This finding implied that the drug-drug interactions had a complementary role in the development of SDDS via ZIF-8. From the metadynamics simulation, it was found that the geometry of the drugs is a determining factor in an efficient co-loading SDDS.
Collapse
Affiliation(s)
- Ahmad Haghi
- Department of Chemistry, University of Birjand Birjand Iran
| | - Heidar Raissi
- Department of Chemistry, University of Birjand Birjand Iran
| | | | | |
Collapse
|
36
|
Yaneva Z, Ivanova D. Catechins within the Biopolymer Matrix-Design Concepts and Bioactivity Prospects. Antioxidants (Basel) 2020; 9:E1180. [PMID: 33256098 PMCID: PMC7761086 DOI: 10.3390/antiox9121180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Epidemiological studies and clinical investigations proposed that catechins extracts alone may not provide a sufficient level of bioactivities and promising therapeutic effects to achieve health benefits due to a number of constraints related to poor oral absorption, limited bioavailability, sensitivity to oxidation, etc. Modern scientific studies have reported numerous techniques for the design of micro- and nano-bio-delivery systems as novel and promising strategies to overcome these obstacles and to enhance catechins' therapeutic activity. The objective assessment of their benefits, however, requires a critical comparative estimation of the advantages and disadvantages of the designed catechins-biocarrier systems, their biological activities and safety administration aspects. In this respect, the present review objectively outlines, compares and assesses the recent advances related to newly developed design concepts of catechins' encapsulation into various biopolymer carriers and their release behaviour, with a special emphasis on the specific physiological biofunctionalities of the innovative bioflavonoid/biopolymer delivery systems.
Collapse
Affiliation(s)
- Zvezdelina Yaneva
- Chemistry Unit, Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, Students Campus, 6000 Stara Zagora, Bulgaria;
| | | |
Collapse
|
37
|
Zhang M, Yang R, Zhou Z, Li C, Liu Y, Li W, Pan J, Sun M, Qian C. Tissue-Specific Regulation of Reactive Oxygen Species by an ATP-Responsive Nanoregulator Enhances Anticancer Efficacy and Reduces Anthracycline-Induced Cardiotoxicity. ACS APPLIED BIO MATERIALS 2020; 3:8000-8011. [DOI: 10.1021/acsabm.0c01049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Minghua Zhang
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ruoxi Yang
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhanwei Zhou
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenzi Li
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yadong Liu
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Li
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jiacheng Pan
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
38
|
Peng SL, Lai CH, Chu PY, Hsieh JT, Tseng YC, Chiu SC, Lin YH. Nanotheranostics With the Combination of Improved Targeting, Therapeutic Effects, and Molecular Imaging. Front Bioeng Biotechnol 2020; 8:570490. [PMID: 33042972 PMCID: PMC7523243 DOI: 10.3389/fbioe.2020.570490] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/27/2020] [Indexed: 12/27/2022] Open
Abstract
There is an increasing interest in the design of targeted carrier systems with combined therapeutic and diagnostic modalities. Therapeutic modalities targeting tumors with single ligand-based targeting nanocarriers are insufficient for proficient delivery and for targeting two different surface receptors that are overexpressed in cancer cells. Here, we evaluated an activated nanoparticle delivery system comprising fucoidan/hyaluronic acid to improve therapeutic efficacy. The system comprised polyethylene glycol-gelatin-encapsulated epigallocatechin gallate (EGCG), poly (D,L-lactide-co-glycolide; PLGA), and stable iron oxide nanoparticles (IOs). The latter enables targeting of prostate cancers in their molecular images. We demonstrate the transfer of nanoparticles and their entry into prostate cancer cells through ligand-specific recognition. This system may prove the benefits of drug delivery that enhances the inhibition of cell growth through apoptosis induction. Moreover, the improved targeting of nanotheranostics significantly suppressed orthotopic prostate tumor growth and more accurately targeted tumors compared with systemic combination therapy. In the presence of nanoparticles with iron oxides, the hypointensity of the prostate tumor was visualized on a T2-weignted magnetic resonance image. The diagnostic ability of this system was demonstrated by accumulating fluorescent nanoparticles in the prostate tumor from the in vivo imaging system, computed tomography. It is suggested that theranostic nanoparticles combined with a molecular imaging system can be a promising cancer therapy in the future.
Collapse
Affiliation(s)
- Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Molecular Infectious Disease Research Center, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pei-Yi Chu
- Faculty of Pharmacy, National Yang-Ming University, Taipei, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yen-Chun Tseng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Hsin Lin
- Faculty of Pharmacy, National Yang-Ming University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Institute of Biopharmaceutical Science, Department and Institute of Pharmacology, Center for Advanced Pharmaceutics and Drug Delivery Research, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
39
|
Gao L, Zhao P, Li Y, Yang D, Hu P, Li L, Cheng Y, Yao H. Reversal of P-glycoprotein-mediated multidrug resistance by novel curcumin analogues in paclitaxel-resistant human breast cancer cells. Biochem Cell Biol 2020; 98:484-491. [PMID: 31967866 DOI: 10.1139/bcb-2019-0377] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multidrug resistance (MDR) is a major obstacle for successful cancer chemotherapy, and the main cause of MDR has been attributed to overexpression of P-glycoprotein (P-gp). In this present study, four P-gp modulators (E,E)-4,6-bis(styryl)-2-(substituted amino)-pyrimidines were evaluated for their activity in a breast cancer cell line overexpressing P-gp (LCC6MDR). The four modulators displayed significantly better P-gp modulating activity compared with the positive control verapamil (RF = 5.4), with a relative fold (RF) increase in activity ranging from 33.3 to 86.0. In contrast to compounds a and c that exhibited lower cytotoxicity, compounds b and d were nontoxic towards both cancer cells and normal cells, with IC50 values greater than 100 μmol/L. The qRT-PCR results demonstrated that after exposure to 2 μmol/L of compounds a, b, c, and d, the mRNA expression level of MDR1 in LCC6MDR cells decreased to 45%, 50%, 38%, and 51%, respectively. However, the Western-blot results indicated that compound c could reverse P-gp mediated MDR, but not via decreases in protein expression. DOX and Rh123 accumulation and efflux results further confirmed that the reversal of MDR activity happens via inhibition of P-gp efflux and increases in intracellular drug accumulation. These results demonstrated that compound c has low toxicity and is an efficient P-gp modulator, highlighting its potential as a promising candidate for P-gp-mediated reversal of MDR.
Collapse
Affiliation(s)
- Lei Gao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People’s Hospital Affiliated to Shandong University, Liaocheng, 252000, China
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, 250000, China
| | - Peiran Zhao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People’s Hospital Affiliated to Shandong University, Liaocheng, 252000, China
| | - Yang Li
- Zhong Yuan Academy of Biological Medicine, Liaocheng People’s Hospital Affiliated to Shandong University, Liaocheng, 252000, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People’s Hospital Affiliated to Shandong University, Liaocheng, 252000, China
| | - Ping Hu
- Zhong Yuan Academy of Biological Medicine, Liaocheng People’s Hospital Affiliated to Shandong University, Liaocheng, 252000, China
| | - Lianzhi Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, 252059, China
| | - Yufeng Cheng
- Department of Radiotherapy, Qilu Hospital of Shandong University, Jinan, 250000, China
| | - Hengchen Yao
- Department of Cardiology, Liaocheng People’s Hospital Affiliated to Shandong University & Shandong First Medical University, Liaocheng, 252000, China
| |
Collapse
|
40
|
Chakouri N, Farah C, Matecki S, Amedro P, Vincenti M, Saumet L, Vergely L, Sirvent N, Lacampagne A, Cazorla O. Screening for in-vivo regional contractile defaults to predict the delayed Doxorubicin Cardiotoxicity in Juvenile Rat. Am J Cancer Res 2020; 10:8130-8142. [PMID: 32724462 PMCID: PMC7381739 DOI: 10.7150/thno.47407] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Anthracyclines are key chemotherapeutic agents used in various adult and pediatric cancers, however, their clinical use is limited due to possible congestive heart failure (HF) caused by acute and irreversible cardiotoxicity. Currently, there is no method to predict the future development of the HF in these patients. In order to identify early biomarkers to predict anthracycline cardiotoxicity in long-term survivors of childhood cancer, this longitudinal study aimed to analyze early and late in-vivo regional myocardial anthracycline-induced cardiotoxicity, related to in-vitro cardiac myocytes dysfunction, in a juvenile rat model. Methods: Young male Wistar rats (4 weeks-old) were treated with different cumulative doses of doxorubicin (7.5, 10 or 12.5 mg/kg) or NaCl (0.9%) once a week for 6 weeks by intravenous injection. Cardiac function was evaluated in-vivo by conventional (left ventricular ejection fraction, LVEF) and regional two-dimensional (2D) speckle tracking echocardiography over the 4 months after the last injection. The animals were assigned to preserved (pEF) or reduced EF (rEF) groups at the end of the protocol and were compared to controls. Results: We observed a preferential contractile dysfunction of the base of the heart, further altered in the posterior segment, even in pEF group. The first regional alterations appeared 1 month after chemotherapy. Functional investigation of cardiomyocytes isolated from the LV base 1 month after doxorubicin treatment showed that early in-vivo contractile alterations were associated with both decreased myofilament Ca2+ sensitivity and length-dependent activation. Changes in post-translational modifications (phosphorylation; S-glutathionylation) and protein degradation of the cardiac myosin binding protein-C may contribute to these alterations. Conclusion: Our data suggest that screening of the contractile defaults of the base of the heart by regional 2D strain echocardiography is useful to detect subclinical myocardial dysfunction prior to the development of delayed anthracycline-induced cardiomyopathy in pediatric onco-cardiology.
Collapse
|
41
|
Guo Z, Yang Y, Shu Y, Qiao L, Peng M, Wang Z. Stimulus-responsive tea polyphenols as nanocarrier for selective intracellular drug delivery. J Biomater Appl 2020; 35:149-157. [PMID: 32390570 DOI: 10.1177/0885328220924539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nanodrug delivery systems have been widely researched to achieve efficient antitumor drug delivery. However, the controlled drug delivery at tumor cells remains the main challenge for antitumor therapy. Herein, a pH and reduction-responsive nanocarrier based on green tea polyphenols was employed as a smart excipient for chemotherapy drug delivery. Paclitaxel, as a chemotherapy drug, was loaded in the nanocarrier, noted as green tea polyphenol/paclitaxel. The green tea polyphenol/paclitaxel kept constant diameter at physiological condition (i.e. pH 7.4), while gradually enlarged at acid environment (pH = 5.5) and the reductive environment. The in vitro paclitaxel release results indicated that the release of paclitaxel from the green tea polyphenol/paclitaxel at pH 7.4 was slow, whereas obviously accelerated at the acid environment (pH = 5.5) and the reductive environment. The in vitro antitumor assay showed more efficient tumor cells inhibition of green tea polyphenol/paclitaxel than free paclitaxel. Meanwhile, due to the proper size (∼100 nm), green tea polyphenol/paclitaxel could effectively accumulate at tumor sites. In the in vivo mice bearing A549 xenograft mouse models, green tea polyphenol/paclitaxel exhibited satisfactory antitumor effect and depressed system toxicity when compared with free paclitaxel, owing to the enhanced paclitaxel accumulation and controlled paclitaxel release in the tumor cells. With simple compositions and satisfactory antitumor effects, this green tea polyphenol-based nanocarrier can be a promising nanodrug delivery system for the therapy of cancers.
Collapse
Affiliation(s)
- Zhiheng Guo
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of the Center for Reproductive Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yang Shu
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, China
| | - Li Qiao
- Department of Gynecological II, The First Hospital of Jilin University, Changchun, China
| | - Min Peng
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, China
| | - Zhenpeng Wang
- Department of Gynecologic Oncologic, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Li S, Li H, Xu X, Saw PE, Zhang L. Nanocarrier-mediated antioxidant delivery for liver diseases. Theranostics 2020; 10:1262-1280. [PMID: 31938064 PMCID: PMC6956819 DOI: 10.7150/thno.38834] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Liver is the principal detoxifying organ and metabolizes various compounds that produce free radicals (FR) constantly. To maintain the oxidative/antioxidative balance in the liver, antioxidants would scavenge FR by preventing tissue damage through FR formation, scavenging, or by enhancing their decomposition. The disruption of this balance therefore leads to oxidative stress and in turn leads to the onset of various diseases. Supplying the liver with exogeneous antioxidants is an effective way to recreate the oxidative/antioxidative balance in the liver homeostasis. Nevertheless, due to the short half-life and instability of antioxidants in circulation, the methodology for delivering antioxidants to the liver needs to be improved. Nanocarrier mediated delivery of antioxidants proved to be an ingenious way to safely and efficiently deliver a high payload of antioxidants into the liver for circumventing liver diseases. The objective of this review is to provide an overview of the role of reactive oxygen species (oxidant) and ROS scavengers (antioxidant) in liver diseases. Subsequently, current nanocarrier mediated antioxidant delivery methods for liver diseases are discussed.
Collapse
Affiliation(s)
- Senlin Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Huiru Li
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| | - Lei Zhang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, People's Republic of China
| |
Collapse
|
43
|
Wang Y, Jiang Z, Yuan B, Tian Y, Xiang L, Li Y, Yang Y, Li J, Wu A. A Y 1 receptor ligand synergized with a P-glycoprotein inhibitor improves the therapeutic efficacy of multidrug resistant breast cancer. Biomater Sci 2019; 7:4748-4757. [PMID: 31508613 DOI: 10.1039/c9bm00337a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Multidrug resistance (MDR) is one of the main reasons for the inefficiency of cancer chemotherapy. As a consequence of MDR, the expression level of membrane proteins might be changed, which can thus be used to develop a novel strategy for its treatment. Based on the high overexpression of Y1 receptor (Y1R) protein and P-glycoprotein (P-gp) in the multidrug resistant breast cancer cell line, a selective Y1R ligand [Asn6, Pro34]-NPY (AP) was employed to stabilize the chemotherapeutic drug doxorubicin (DOX) and P-gp inhibitor tariquidar (Tar) co-loaded nanomicelles at the physiological level. This also improved the targeted delivery of DOX and Tar into MCF-7/ADR cells. Co-delivered Tar further impedes the efflux of DOX and enhances its accumulation in the nuclei of drug resistant cancer cells, thereby inducing significant inhibition of cell growth. The synergistic effect of AP and Tar generates an excellent in vivo tumor targeting and antitumor efficacy of DOX with prolonged survival and minimized side effects, especially for liver metastasis. In general, Y1R as a novel target site and its selective ligand AP synergized with the P-gp inhibitor can be used for a more precise MDR breast cancer treatment.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Screening Assays, Antitumor
- Female
- Humans
- Ligands
- MCF-7 Cells
- Mammary Neoplasms, Experimental/diagnostic imaging
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Micelles
- Nanoparticles/chemistry
- Optical Imaging
- Quinolines/chemistry
- Quinolines/pharmacology
- Receptors, Neuropeptide Y/antagonists & inhibitors
- Receptors, Neuropeptide Y/metabolism
Collapse
Affiliation(s)
- Yinjie Wang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenqi Jiang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Yuan
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Yuchen Tian
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Lingchao Xiang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Yanying Li
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong Yang
- Department of Clinical Laboratory, the Affiliated Hospital of Medical School of Ningbo University, Ningbo University School of Medicine, Ningbo, 315010, China
| | - Juan Li
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P.R. China.
| |
Collapse
|
44
|
Zhang W, Zhang W, Sun L, Xiang L, Lai X, Li Q, Sun S. The effects and mechanisms of epigallocatechin-3-gallate on reversing multidrug resistance in cancer. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.09.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Zhang L, Wu P, Zhang L, SreeHarsha N, Mishra A, Su X. Ameliorative effect of rosiglitazone, a peroxisome proliferator gamma agonist on adriamycin-induced cardio toxicity via suppressing oxidative stress and apoptosis. IUBMB Life 2019; 72:607-615. [PMID: 31660680 DOI: 10.1002/iub.2190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 10/01/2019] [Indexed: 12/19/2022]
Abstract
We investigated the rosiglitazone (RSG) effect on adriamycin (ADM)-induced cardio toxicity in experimental animals. Forty adult Wistar male rats were separated into four groups as follows: normal control; RSG (10 mg/kg)-treated; ADM (10 mg/kg)-administered; and ADM (10 mg/kg) + RSG (10 mg/kg)-treated. Serum lipid level, different biochemical biomarkers, histological analysis, and nuclear factor erythroid 2-related factor/heme oxygenase-1 (Nrf2/HO-1), Caspase 3, B-cell lymphoma 2 (Bcl-2), and Bax gene expression were assessed in serum and cardiac tissue samples. Our results show that RSG treatment in ADM-administered animals significantly diminished low-density lipoprotein cholesterol, triglyceride, and total cholesterol, and increases high-density lipoprotein cholesterol (HDL-c) in comparison with the ADM group. RSG treatment reduced the effect of ADM administration on cardiac dysfunction markers such as cardiac troponin T Creatine Kinase-MB, aspartate aminotransferase, and lactate dehydrogenase, showing the amelioration of cardio toxicity in ADM-administered rats. Additionally, RSG treatment significantly decreased the level of malondialdehyde and nitric oxide in cardiovascular tissue. RSG-treated rats in combination with ADM likewise showed a significant increase in reduced glutathione, superoxide dismutase, catalase content, and the activity of glutathione peroxidase (GPx) as compared with ADM group. Moreover, RSG treatment in ADM rats significantly increased an Nrf2 and HO-1 expression in comparison with ADM group. While in apoptosis parameters, RSG treatment in ADM rats significantly diminished a cleaved caspase-3 and Bax expression as well as expanded Bcl-2 expression when contrasted with ADM group of rats. In conclusion, RSG is capable of protecting heart toxicity in ADM-treated animals through defensive effects on oxidative stress and biochemical markers.
Collapse
Affiliation(s)
- Lingling Zhang
- Department of Cardiology, Binzhou People's Hospital, Binzhou, China
| | - Ping Wu
- Department Rear-Service, Binzhou People's Hospital, Binzhou, China
| | - Luyan Zhang
- Department of Oncology, Binzhou People's Hospital, Binzhou, China
| | - Nagaraja SreeHarsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Anurag Mishra
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
| | - Xinyou Su
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
46
|
Ren C, Gao Y, Guan Y, Wang Z, Yang L, Gao J, Fan H, Liu J. Carrier-Free Supramolecular Hydrogel Composed of Dual Drugs for Conquering Drug Resistance. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33706-33715. [PMID: 31466443 DOI: 10.1021/acsami.9b12530] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The resistance of tumor cells to anticancer drugs has become one of the principal causes of the failure in clinical chemotherapy. To overcome this issue, developing feasible drug delivery systems for effective cancer therapy is urgently needed. In this work, we construct an amphiphilic drug self-delivery system consisting of Taxol and tyroservatide (YSV) to overcome drug resistance. The carrier-free supramolecular hydrogel composed of nanofibers is formed by the involved ester bond self-hydrolysis process, which has high drug loading efficiency and facilitates the delivery of both the hydrophobic Taxol and hydrophilic YSV. Because of the dual inhibitory function of YSV on histone deacetylase and P-glycoprotein, an improved combinational anticancer effect of the molecule against drug-resistant tumor cells in vitro is achieved. Furthermore, the designed drug self-delivery system exhibited enhanced antitumor efficiency and favorable biocompatibility in vivo when administered by tail vein injection. Our study provides a new strategy for fabricating a carrier-free supramolecular hydrogel to overcome drug resistance, which might open up an alternative avenue for the tumor combinational therapy.
Collapse
Affiliation(s)
- Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Yang Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Yong Guan
- Department of Pediatric Urology , Tianjin Children's Hospital , Tianjin 300134 , China
| | - Zhongyan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Lijun Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Jie Gao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Huirong Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192 , China
| |
Collapse
|
47
|
Aiello P, Consalvi S, Poce G, Raguzzini A, Toti E, Palmery M, Biava M, Bernardi M, Kamal MA, Perry G, Peluso I. Dietary flavonoids: Nano delivery and nanoparticles for cancer therapy. Semin Cancer Biol 2019; 69:150-165. [PMID: 31454670 DOI: 10.1016/j.semcancer.2019.08.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/08/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
Abstract
Application of nanotechnologies to cancer therapy might increase solubility and/or bioavailability of bioactive compounds of natural or synthetic origin and offers other potential benefits in cancer therapy, including selective targeting. In the present review we aim to evaluate in vivo studies on the anticancer activity of nanoparticles (NPs) obtained from food-derived flavonoids. From a systematic search a total of 60 studies were identified. Most of the studies involved the flavanol epigallocatechin-3-O-gallate and the flavonol quercetin, in both delivery and co-delivery (with anti-cancer drugs) systems. Moreover, some studies investigated the effects of other flavonoids, such as anthocyanins aglycones anthocyanidins, flavanones, flavones and isoflavonoids. NPs inhibited tumor growth in both xenograft and chemical-induced animal models of cancerogenesis. Encapsulation improved bioavailability and/or reduced toxicity of both flavonoids and/or co-delivered drugs, such as doxorubicin, docetaxel, paclitaxel, honokiol and vincristine. Moreover, flavonoids have been successfully applied in molecular targeted nanosystems. Selectivity for cancer cells involves pH- and/or reactive oxygen species-mediated mechanisms. Furthermore, flavonoids are good candidates as drug delivery for anticancer drugs in green synthesis systems. In conclusion, although human studies are needed, NPs obtained from food-derived flavonoids have promising anticancer effects in vivo.
Collapse
Affiliation(s)
- Paola Aiello
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy; Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - Sara Consalvi
- Department of Chemistry and Drug Technologies, University "La Sapienza", Rome, Italy
| | - Giovanna Poce
- Department of Chemistry and Drug Technologies, University "La Sapienza", Rome, Italy
| | - Anna Raguzzini
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Elisabetta Toti
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Maura Palmery
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy
| | - Mariangela Biava
- Department of Chemistry and Drug Technologies, University "La Sapienza", Rome, Italy
| | - Marco Bernardi
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW, 2770, Australia; Novel Global Community Educational Foundation, Australia
| | - George Perry
- Department of Biology, University of Texas at San Antonio, TX, USA.
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy.
| |
Collapse
|
48
|
Li Y, Zhang T, Liu Q, He J. PEG-Derivatized Dual-Functional Nanomicelles for Improved Cancer Therapy. Front Pharmacol 2019; 10:808. [PMID: 31379579 PMCID: PMC6659352 DOI: 10.3389/fphar.2019.00808] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/24/2019] [Indexed: 02/05/2023] Open
Abstract
Polymeric micelles have attracted considerable attention for effective delivery of poorly water-soluble cancer drugs. Polyethylene glycol (PEG), which has been approved for human use by the US Food and Drug Administration, is the most commonly used hydrophilic component of polymeric micelles because it is biocompatible and biodegradable. One disadvantage of traditional polymeric micelles is that they include a large amount of inert carrier materials, which do not contribute to therapeutic activity but increase cost and toxicity risk. A better alternative may be "dual-functional" micellar carriers, in which the hydrophobic carrier material (conjugated to PEG) has intrinsic therapeutic activity that complements, or even synergizes with, the antitumor activity of the drug cargo. This review summarizes recent progress in the development of PEG-derivatized dual-functional nanomicelles and surveys the evidence of their feasibility and promise for cancer therapy.
Collapse
Affiliation(s)
- Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Zhang
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, China.,Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
49
|
He J, Xu L, Yang L, Sun C. Anti-oxidative effects of catechins and theaflavins on glutamate-induced HT22 cell damage. RSC Adv 2019; 9:21418-21428. [PMID: 35521307 PMCID: PMC9066190 DOI: 10.1039/c9ra02721a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/30/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Glutamate is an excitatory neurotransmitter that is involved in cell stress caused by oxidation. Polyphenolic compounds display various potential neuroprotective properties due to their ability to donate electrons and hydrogen atoms. Method: In this study, we evaluate the protective effect towards glutamate-induced HT22 cell damage. Two families of polyphenolic compounds are investigated, including the monomer polyphenol catechins, as well as the dimerized theaflavins. The cell apoptosis and intercellular ROS production are quantified by flow cytometry, and the protective mechanism is evaluated by quantifying the expression of cell apoptosis and energy related proteins. Result: Both sets of compounds protect cells against glutamate-induced oxidative stress, partially restore the cell viability, and prevent cells from apoptosis via bcl-2 and bax regulation, and attenuate intercellular ROS production. We demonstrate here that the protective effect is mediated by multiple factors, including reducing intracellular Ca2+ concentration, increasing glutathione level and related enzyme activity. Thus, the phosphorylation of AMP-activated protein kinase (AMPK) and extracellular signal-regulated kinase (ERK) show inverse correlation of activity after catechins and theaflavins stimulation. Conclusion: These results suggest both catechins and theaflavins compounds protect cells from glutamate-induced damage via cell apoptosis-related proteins and indirect regulation of cellular energy enzymes. These natural sourced antioxidants provide potential therapeutic agents for glutamate accumulation and toxicity related diseases.
Collapse
Affiliation(s)
- Jinting He
- Department of Neurology, China-Japan Union Hospital, Jilin University Changchun China 130031
| | - Lei Xu
- Department of Neurology, China-Japan Union Hospital, Jilin University Changchun China 130031
| | - Le Yang
- People's Hospital of Jilin Province Changchun China 130021
| | - Caixia Sun
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital, Jilin University 126 Xiantai St, Erdao Qu Changchun Jilin China 130031
| |
Collapse
|
50
|
Shan L, Gao G, Wang W, Tang W, Wang Z, Yang Z, Fan W, Zhu G, Zhai K, Jacobson O, Dai Y, Chen X. Self-assembled green tea polyphenol-based coordination nanomaterials to improve chemotherapy efficacy by inhibition of carbonyl reductase 1. Biomaterials 2019; 210:62-69. [PMID: 31075724 PMCID: PMC6521851 DOI: 10.1016/j.biomaterials.2019.04.032] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/17/2019] [Accepted: 04/28/2019] [Indexed: 11/19/2022]
Abstract
Nanomedicine has become a promising approach to improve cancer chemotherapy. It remains a major challenge how to enhance anti-drug efficacy and reduce side effects of anti-cancer drugs. Herein, we report a self-assembled nanoplatform (FDEP NPs) by integration of doxorubicin (DOX) and epigallocatechin-3-O-gallate (EGCG) with the help of coordination between Fe3+ ions and polyphenols. The EGCG from FDEP NPs could inhibit the expression of carbonyl reductase 1 (CBR1) protein and thereby inhibit the doxorubicinol (DOXOL) generation from DOX both in vitro and in vivo, thus the efficacy of DOX to cancerous cells is improved significantly. More importantly, the FDEP NPs could reduce cardiac toxicity and the DOX mediated toxicity to blood cells due to the repression of DOXOL production. Moreover, the blood half-life of FDEP NPs is longer than 23 h as determined by positron emission tomography (PET) imaging of biodistribution of radiolabelled NPs and HPLC measurement of plasma level of DOX, ensuring high tumor accumulation of FDEP NPs by enhanced permeability and retention (EPR) effect. The FDEP NPs also exhibited much improved antitumor effect over free drugs. Our work sheds new light on the engineering of nanomaterials for combination chemotherapy and may find unique clinical applications in the near future.
Collapse
Affiliation(s)
- Lingling Shan
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, PR China; Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Guizhen Gao
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Weiwei Wang
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Wei Tang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Zhen Yang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Guizhi Zhu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Kefeng Zhai
- Institute of Pharmaceutical Biotechnology, School of Biology and Food Engineering, Suzhou University, Suzhou 234000, PR China
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States
| | - Yunlu Dai
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, PR China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, PR China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, United States.
| |
Collapse
|