51
|
Yu X, Li X, Chen Q, Wang S, Xu R, He Y, Qin X, Zhang J, Yang W, Shi L, Lu L, Zheng Y, Pang Z, Peng S. High Intensity Focused Ultrasound-Driven Nanomotor for Effective Ferroptosis-Immunotherapy of TNBC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305546. [PMID: 38342612 PMCID: PMC11022700 DOI: 10.1002/advs.202305546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/14/2024] [Indexed: 02/13/2024]
Abstract
The heterogeneity of triple-negative breast cancers (TNBC) remains challenging for various treatments. Ferroptosis, a recently identified form of cell death resulting from the unrestrained peroxidation of phospholipids, represents a potential vulnerability in TNBC. In this study, a high intensity focused ultrasound (HIFU)-driven nanomotor is developed for effective therapy of TNBC through induction of ferroptosis. Through bioinformatics analysis of typical ferroptosis-associated genes in the FUSCCTNBC dataset, gambogic acid is identified as a promising ferroptosis drug and loaded it into the nanomotor. It is found that the rapid motion of nanomotors propelled by HIFU significantly enhanced tumor accumulation and penetration. More importantly, HIFU not only actuated nanomotors to trigger effective ferroptosis of TNBC cells, but also drove nanomotors to activate ferroptosis-mediated antitumor immunity in primary and metastatic TNBC models, resulting in effective tumor regression and prevention of metastases. Overall, HIFU-driven nanomotors show great potential for ferroptosis-immunotherapy of TNBC.
Collapse
Affiliation(s)
- Xiangrong Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| | - Xuejing Li
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Qingwang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome InstituteFudan University2005 Songhu RoadShanghai200438P.R. China
| | - Siyu Wang
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Ruizhe Xu
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Ying He
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Xifeng Qin
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical NeurobiologyFudan University12 Wulumuqi Middle RoadShanghai200040China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular ScienceFudan UniversityShanghai200433China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome InstituteFudan University2005 Songhu RoadShanghai200438P.R. China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| | - Yuanting Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome InstituteFudan University2005 Songhu RoadShanghai200438P.R. China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, School of PharmacyFudan UniversityShanghai201203P.R. China
| | - Shaojun Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| |
Collapse
|
52
|
Wang Y, Wang L, Chang H, Shen Q, Zhang S, Sun S, Liu Y, Zheng J, Liu H. Enhancing anti-tumor therapy with agmatine-cholesterol conjugate liposomes: in vitro and in vivo evidence. Drug Deliv Transl Res 2024; 14:788-801. [PMID: 37755673 DOI: 10.1007/s13346-023-01433-5] [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] [Accepted: 09/03/2023] [Indexed: 09/28/2023]
Abstract
In this study, we synthesized a novel compound, agmatine-cholesterol conjugate (AG-Chol), to enhance the anti-tumor activity of drug-loaded liposomes. We replaced cholesterol with AG-Chol in preparing doxorubicin hydrochloride (DOX) liposomes by using an active loading method for DOX. We assessed the physical and chemical properties of the resulting AG-Liposomes and evaluated their efficacy in vitro and in vivo. The results showed that AG-Liposomes were stable with high encapsulation efficiency. Compared with the control liposomes, AG-Liposomes exhibited a slower drug release rate in the release medium at pH 6.8. The in vitro cell experiments demonstrated that AG-Liposomes had higher tumor cell uptake rate, stronger migration inhibition rate, higher apoptosis rate, better anti-clonogenic ability, and higher lysosome escape ability than the control liposomes. In vivo distribution results demonstrate that liposomes prepared with AG-Chol instead of cholesterol can significantly enhance their tumor targeting abilities and reduce their distribution to non-targeted sites. In vivo tumor suppression experiments showed that AG-Liposomes had a higher tumor suppression rate than the control liposomes without causing apparent toxicity to normal tissues, as evidenced by histological staining. Therefore, substituting cholesterol with AG-Chol in the preparation of liposomes can result in enhanced lysosome escape, improved tumor targeting, and increased efficacy of anti-tumor drugs.
Collapse
Affiliation(s)
- Yanzhi Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China.
| | - Linchao Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China
- Jining No. 1 People's Hospital, Jining, China
| | - Hanyue Chang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China
| | - Qing Shen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China
| | - Sai Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China
| | - Shanshan Sun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China
| | - Ying Liu
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiaxin Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China.
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Ministry of Education of China, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
53
|
Arafat M, Sakkal M, Beiram R, AbuRuz S. Nanomedicines: Emerging Platforms in Smart Chemotherapy Treatment-A Recent Review. Pharmaceuticals (Basel) 2024; 17:315. [PMID: 38543101 PMCID: PMC10974155 DOI: 10.3390/ph17030315] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 01/06/2025] Open
Abstract
Cancer continues to pose one of the most critical challenges in global healthcare. Despite the wide array of existing cancer drugs, the primary obstacle remains in selectively targeting and eliminating cancer cells while minimizing damage to healthy ones, thereby reducing treatment side effects. The revolutionary approach of utilizing nanomaterials for delivering cancer therapeutic agents has significantly enhanced the efficacy and safety of chemotherapeutic drugs. This crucial shift is attributed to the unique properties of nanomaterials, enabling nanocarriers to transport therapeutic agents to tumor sites in both passive and active modes, while minimizing drug elimination from delivery systems. Furthermore, these nanocarriers can be designed to respond to internal or external stimuli, thus facilitating controlled drug release. However, the production of nanomedications for cancer therapy encounters various challenges that can impede progress in this field. This review aims to provide a comprehensive overview of the current state of nanomedication in cancer treatment. It explores a variety of nanomaterials, focusing on their unique properties that are crucial for overcoming the limitations of conventional chemotherapy. Additionally, the review delves into the properties and functionalities of nanocarriers, highlighting their significant impact on the evolution of nanomedicine. It also critically assesses recent advancements in drug delivery systems, covering a range of innovative delivery methodologies. Finally, the review succinctly addresses the challenges encountered in developing nanomedications, offering insightful perspectives to guide future research in this field.
Collapse
Affiliation(s)
- Mosab Arafat
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (M.A.)
| | - Molham Sakkal
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (M.A.)
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Salahdein AbuRuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
54
|
Thirumurugan S, Dash P, Lin YC, Sakthivel R, Sun YS, Lin CP, Wang AN, Liu X, Dhawan U, Tung CW, Chung RJ. Synergistic effect of photothermal and magnetic hyperthermia for in situ activation of Fenton reaction in tumor microenvironment for chemodynamic therapy. BIOMATERIALS ADVANCES 2024; 157:213724. [PMID: 38134729 DOI: 10.1016/j.bioadv.2023.213724] [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: 08/31/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
Traditional cancer treatments are ineffective and cause severe adverse effects. Thus, the development of chemodynamic therapy (CDT) has the potential for in situ catalysis of endogenous molecules into highly toxic species, which would then effectively destroy cancer cells. However, the shortage of high-performance nanomaterials hinders the broad clinical application of this approach. In present study, an effective therapeutic platform was developed using a simple hydrothermal method for the in-situ activation of the Fenton reaction within the tumor microenvironment (TME) to generate substantial quantities of •OH and ultimately destroy cancer cells, which could be further synergistically increased by photothermal therapy (PHT) and magnetic hyperthermia (MHT) aided by FeMoO4 nanorods (NRs). The produced FeMoO4 NRs were used as MHT/PHT and Fenton catalysts. The photothermal conversion efficiency of the FeMoO4 NRs was 31.75 %. In vitro and \ experiments demonstrated that the synergistic combination of MHT/PHT/CDT notably improved anticancer efficacy. This work reveals the significant efficacy of CDT aided by both photothermal and magnetic hyperthermia and offers a feasible strategy for the use of iron-based nanoparticles in the field of biomedical applications.
Collapse
Affiliation(s)
- Senthilkumar Thirumurugan
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan
| | - Pranjyan Dash
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan
| | - Yu-Chien Lin
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan
| | - Rajalakshmi Sakthivel
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan
| | - Ying-Sui Sun
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | | | - Xinke Liu
- College of Materials Science and Engineering, Chinese Engineering and Research Institute of Microelectronics, Shenzhen University, Shenzhen 518060, China; Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Udesh Dhawan
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G116EW, UK
| | - Ching-Wei Tung
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City 243303, Taiwan.
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan; High-value Biomaterials Research and Commercialization Center, National Taipei University of Technology (Taipei Tech), Taipei 106344, Taiwan.
| |
Collapse
|
55
|
Jiang Y, Chen H, Lin T, Zhang C, Shen J, Chen J, Zhao Y, Xu W, Wang G, Huang P. Ultrasound-activated prodrug-loaded liposome for efficient cancer targeting therapy without chemotherapy-induced side effects. J Nanobiotechnology 2024; 22:2. [PMID: 38169390 PMCID: PMC10763105 DOI: 10.1186/s12951-023-02195-5] [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: 09/12/2023] [Accepted: 11/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Off-targeted distribution of chemotherapeutic drugs causes severe side effects, further leading to poor prognosis and patient compliance. Ligand/receptor-mediated targeted drug delivery can improve drug accumulation in the tumor but it always attenuated by protein corona barriers. RESULTS To address these problems, a radically different strategy is proposed that can leave the off-targeted drugs inactive but activate the tumor-distributed drugs for cancer-targeting therapy in a tumor microenvironment-independent manner. The feasibility and effectiveness of this strategy is demonstrated by developing an ultrasound (US)-activated prodrug-loaded liposome (CPBSN38L) comprising the sonosensitizer chlorin e6 (Ce6)-modified lipids and the prodrug of pinacol boronic ester-conjugated SN38 (PBSN38). Once CPBSN38L is accumulated in the tumor and internalized into the cancer cells, under US irradiation, the sonosensitizer Ce6 rapidly induces extensive production of intracellular reactive oxygen species (ROS), thereby initiating a cascade amplified ROS-responsive activation of PBSN38 to release the active SN38 for inducing cell apoptosis. If some of the injected CPBSN38L is distributed into normal tissues, the inactive PBSN38 exerts no pharmacological activity on normal cells. CPBSN38L exhibited strong anticancer activity in multiple murine tumor models of colon adenocarcinoma and hepatocellular carcinoma with no chemotherapy-induced side effects, compared with the standard first-line anticancer drugs irinotecan and topotecan. CONCLUSIONS This study established a side-effect-evitable, universal, and feasible strategy for cancer-targeting therapy.
Collapse
Affiliation(s)
- Yifan Jiang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hongjian Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Tao Lin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiaxin Shen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanan Zhao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Wen Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
56
|
Li Y, Wei C, Yan J, Li F, Chen B, Sun Y, Luo K, He B, Liang Y. The application of nanoparticles based on ferroptosis in cancer therapy. J Mater Chem B 2024; 12:413-435. [PMID: 38112639 DOI: 10.1039/d3tb02308g] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Ferroptosis is a new form of non-apoptotic programmed cell death. Due to its effectiveness in cancer treatment, there are increasing studies on the application of nanoparticles based on ferroptosis in cancer therapy. In this paper, we present a summary of the latest progress in nanoparticles based on ferroptosis for effective tumor therapy. We also describe the combined treatment of ferroptosis with other therapies, including chemotherapy, radiotherapy, phototherapy, immunotherapy, and gene therapy. This summary of drug delivery systems based on ferroptosis aims to provide a basis and inspire opinions for researchers concentrating on exploring this field. Finally, we present some prospects and challenges for the application of nanotherapies to clinical treatment by promoting ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yifei Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Chen Wei
- Department of Pharmacy, Qingdao Women and Children's Hospital, Qingdao 266034, China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Fashun Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Bohan Chen
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| |
Collapse
|
57
|
Sharma V, Arora A, Bansal S, Semwal A, Sharma M, Aggarwal A. Role of bio-flavonols and their derivatives in improving mitochondrial dysfunctions associated with pancreatic tumorigenesis. Cell Biochem Funct 2024; 42:e3920. [PMID: 38269510 DOI: 10.1002/cbf.3920] [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: 09/06/2023] [Revised: 11/30/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024]
Abstract
Mitochondria, a cellular metabolic center, efficiently fulfill cellular energy needs and regulate crucial metabolic processes, including cellular proliferation, differentiation, apoptosis, and generation of reactive oxygen species. Alteration in the mitochondrial functions leads to metabolic imbalances and altered extracellular matrix dynamics in the host, utilized by solid tumors like pancreatic cancer (PC) to get energy benefits for fast-growing cancer cells. PC is highly heterogeneous and remains unidentified for a longer time because of its complex pathophysiology, retroperitoneal position, and lack of efficient diagnostic approaches, which is the foremost reason for accounting for the seventh leading cause of cancer-related deaths worldwide. PC cells often respond poorly to current therapeutics because of dense stromal barriers in the pancreatic tumor microenvironment, which limit the drug delivery and distribution of antitumor immune cell populations. As an alternative approach, various natural compounds like flavonoids are reported to possess potent antioxidant and anticancerous properties and are less toxic than current chemotherapeutic drugs. Therefore, we aim to summarize the current state of knowledge regarding the pharmacological properties of flavonols in PC in this review from the perspective of mitigating mitochondrial dysfunctions associated with cancer cells. Our literature survey indicates that flavonols efficiently regulate cellular metabolism by scavenging reactive oxygen species, mitigating inflammation, and arresting the cell cycle to promote apoptosis in tumor cells via intrinsic mitochondrial pathways. In particular, flavonols proficiently inhibit the cancer-associated proliferation and inflammatory pathways such as EGFR/MAPK, PI3K/Akt, and nuclear factor κB in PC. Overall, this review provides in-depth evidence about the therapeutic potential of flavonols for future anticancer strategies against PC; still, more multidisciplinary human interventional studies are required to dissect their pharmacological effect accurately.
Collapse
Affiliation(s)
- Vinit Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ankita Arora
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sakshi Bansal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ankita Semwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Mayank Sharma
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anjali Aggarwal
- Department of Anatomy, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
58
|
Akaki S, Hosokawa M, Maeda S, Kono Y, Maeda H, Ogawara KI. Efficient Loading into and Controlled Release of Lipophilic Compound from Liposomes by Using Cyclodextrin as Novel Trapping Agent. Biol Pharm Bull 2024; 47:1832-1835. [PMID: 39522976 DOI: 10.1248/bpb.b24-00558] [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: 11/16/2024]
Abstract
Lipid bilayer vesicles, liposomes are representative drug delivery carriers. High encapsulation efficiency and release control of drugs are essential for clinical application of liposomes. For efficient drug loading into liposomes, remote loading method using driving force like transmembrane gradients of pH and ions are utilized. Ions are called as "trapping agents," which are also critical for the controlled release of drugs loaded into liposomes inside. It is difficult to apply ions as trapping agents to various drugs because of limited physicochemical compatibility between drugs and ions. Cyclodextrins (CDs) with hydrophobic cavity can make inclusion complexes with various hydrophobic compounds. Therefore, we aimed to evaluate the potential of CDs as a novel trapping agent using sulfobutylether-β-cyclodextrin (SBE-β-CD) and ibuprofen (IB), a weak acid hydrophobic drug. Encapsulation efficiency of IB in liposomes with pH gradient was approximately 27%, and it was enhanced by intraliposomal SBE-β-CD inclusion in addition to pH gradient, which was SBE-β-CD concentration-dependent. In liposomes with pH gradient, a large fraction of IB was released in a short time. This early-stage rapid IB release was significantly suppressed by the inclusion of SBE-β-CD inside liposomes. Thus, novel remote loading technology by intraliposomal SBE-β-CD enabled the efficient encapsulation of the hydrophobic drug into the aqueous phase of liposomes as well as their controlled release. This technology should be applied to various drugs that can be included into CDs in order to enhance their therapeutic benefits.
Collapse
Affiliation(s)
- Sae Akaki
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University
| | - Mika Hosokawa
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University
| | - Saki Maeda
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University
| | - Yusuke Kono
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University
| | - Hideko Maeda
- Comprehensive Education and Research Center (Support Division), Kobe Pharmaceutical University
| | | |
Collapse
|
59
|
Ebrahimnejad P, Mohammadi Z, Babaei A, Ahmadi M, Amirkhanloo S, Asare-Addo K, Nokhodchid A. Novel Strategies Using Sagacious Targeting for Site-Specific Drug Delivery in Breast Cancer Treatment: Clinical Potential and Applications. Crit Rev Ther Drug Carrier Syst 2024; 41:35-84. [PMID: 37824418 DOI: 10.1615/critrevtherdrugcarriersyst.v41.i1.20] [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: 10/14/2023]
Abstract
For more than a decade, researchers have been working to achieve new strategies and smart targeting drug delivery techniques and technologies to treat breast cancer (BC). Nanotechnology presents a hopeful strategy for targeted drug delivery into the building of new therapeutics using the properties of nanomaterials. Nanoparticles are of high regard in the field of diagnosis and the treatment of cancer. The use of these nanoparticles as an encouraging approach in the treatment of various cancers has drawn the interest of researchers in recent years. In order to achieve the maximum therapeutic effectiveness in the treatment of BC, combination therapy has also been adopted, leading to minimal side effects and thus an enhancement in the quality of life for patients. This review article compares, discusses and criticizes the approaches to treat BC using novel design strategies and smart targeting of site-specific drug delivery systems.
Collapse
Affiliation(s)
- Pedram Ebrahimnejad
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Mohammadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Melika Ahmadi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shervin Amirkhanloo
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kofi Asare-Addo
- Department of Pharmacy, University of Huddersfield, Huddersfield, UK
| | - Ali Nokhodchid
- Lupin Pharmaceutical Research Center, Coral Springs, Florida, USA; Pharmaceutics Research Lab, Arundel Building, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
60
|
Kaushik M, Kumar S, Singh M, Sharma H, Bhowmick M, Bhowmick P, Ashique S, Khatoon H, Pal R, Ansari MA. Bio-inspired Nanomaterials in Cancer Theranostics. NANOTHERANOSTICS FOR DIAGNOSIS AND THERAPY 2024:95-123. [DOI: https:/doi.org/10.1007/978-981-97-3115-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
|
61
|
Ebrahimi N, Manavi MS, Nazari A, Momayezi A, Faghihkhorasani F, Rasool Riyadh Abdulwahid AH, Rezaei-Tazangi F, Kavei M, Rezaei R, Mobarak H, Aref AR, Fang W. Nano-scale delivery systems for siRNA delivery in cancer therapy: New era of gene therapy empowered by nanotechnology. ENVIRONMENTAL RESEARCH 2023; 239:117263. [PMID: 37797672 DOI: 10.1016/j.envres.2023.117263] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
RNA interference (RNAi) is a unique treatment approach used to decrease a disease's excessive gene expression, including cancer. SiRNAs may find and destroy homologous mRNA sequences within the cell thanks to RNAi processes. However, difficulties such poor cellular uptake, off-target effects, and susceptibility to destruction by serum nucleases in the bloodstream restrict the therapeutic potential of siRNAs. Since some years ago, siRNA-based therapies have been in the process of being translated into the clinic. Therefore, the primary emphasis of this work is on sophisticated nanocarriers that aid in the transport of siRNA payloads, their administration in combination with anticancer medications, and their use in the treatment of cancer. The research looks into molecular manifestations, difficulties with siRNA transport, the design and development of siRNA-based delivery methods, and the benefits and drawbacks of various nanocarriers. The trapping of siRNA in endosomes is a challenge for the majority of delivery methods, which affects the therapeutic effectiveness. Numerous techniques for siRNA release, including as pH-responsive release, membrane fusion, the proton sponge effect, and photochemical disruption, have been studied to overcome this problem. The present state of siRNA treatments in clinical trials is also looked at in order to give a thorough and systematic evaluation of siRNA-based medicines for efficient cancer therapy.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Iran
| | | | - Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Amirali Momayezi
- School of Chemical Engineering, Iran University of Science, and Technology, Tehran, Iran
| | | | | | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Mohammed Kavei
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Roya Rezaei
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Halimeh Mobarak
- Clinical Pathologist, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Wei Fang
- Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
62
|
Lee SW, Frankston CM, Kim J. Epigenome editing in cancer: Advances and challenges for potential therapeutic options. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 383:191-230. [PMID: 38359969 DOI: 10.1016/bs.ircmb.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Cancers are diseases caused by genetic and non-genetic environmental factors. Epigenetic alterations, some attributed to non-genetic factors, can lead to cancer development. Epigenetic changes can occur in tumor suppressors or oncogenes, or they may contribute to global cell state changes, making cells abnormal. Recent advances in gene editing technology show potential for cancer treatment. Herein, we will discuss our current knowledge of epigenetic alterations occurring in cancer and epigenetic editing technologies that can be applied to developing therapeutic options.
Collapse
Affiliation(s)
- Seung-Won Lee
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States; Department of Molecular and Medical Genetics, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Connor Mitchell Frankston
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States; Biomedical Engineering Graduate Program, Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Jungsun Kim
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States; Department of Molecular and Medical Genetics, School of Medicine, Oregon Health & Science University, Portland, OR, United States; Cancer Biology Research Program, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
63
|
Santana JEG, Oliveira-Tintino CDDM, Gonçalves Alencar G, Siqueira GM, Sampaio Alves D, Moura TF, Tintino SR, de Menezes IRA, Rodrigues JPV, Gonçalves VBP, Nicolete R, Emran TB, Gonçalves Lima CM, Ahmad SF, Coutinho HDM, da Silva TG. Comparative Antibacterial and Efflux Pump Inhibitory Activity of Isolated Nerolidol, Farnesol, and α-Bisabolol Sesquiterpenes and Their Liposomal Nanoformulations. Molecules 2023; 28:7649. [PMID: 38005371 PMCID: PMC10675182 DOI: 10.3390/molecules28227649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
The efflux systems are considered important mechanisms of bacterial resistance due to their ability to extrude various antibiotics. Several naturally occurring compounds, such as sesquiterpenes, have demonstrated antibacterial activity and the ability to inhibit efflux pumps in resistant strains. Therefore, the objective of this research was to analyze the antibacterial and inhibitory activity of the efflux systems NorA, Tet(K), MsrA, and MepA by sesquiterpenes nerolidol, farnesol, and α-bisabolol, used either individually or in liposomal nanoformulation, against multi-resistant Staphylococcus aureus strains. The methodology consisted of in vitro testing of the ability of sesquiterpenes to reduce the Minimum Inhibitory Concentration (MIC) and enhance the action of antibiotics and ethidium bromide (EtBr) in broth microdilution assays. The following strains were used: S. aureus 1199B carrying the NorA efflux pump, resistant to norfloxacin; IS-58 strain carrying Tet(K), resistant to tetracyclines; RN4220 carrying MsrA, conferring resistance to erythromycin. For the EtBr fluorescence measurement test, K2068 carrying MepA was used. It was observed the individual sesquiterpenes exhibited better antibacterial activity as well as efflux pump inhibition. Farnesol showed the lowest MIC of 16.5 µg/mL against the S. aureus RN4220 strain. Isolated nerolidol stood out for reducing the MIC of EtBr to 5 µg/mL in the 1199B strain, yielding better results than the positive control CCCP, indicating strong evidence of NorA inhibition. The liposome formulations did not show promising results, except for liposome/farnesol, which reduced the MIC of EtBr against 1199B and RN4220. Further research is needed to evaluate the mechanisms of action involved in the inhibition of resistance mechanisms by the tested compounds.
Collapse
Affiliation(s)
| | - Cícera Datiane de Morais Oliveira-Tintino
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Gabriel Gonçalves Alencar
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Gustavo Miguel Siqueira
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Daniel Sampaio Alves
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Talysson Felismino Moura
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Saulo Relison Tintino
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Irwin Rose Alencar de Menezes
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | | | | | - Roberto Nicolete
- Oswaldo Cruz Foundation (Fiocruz Ceará), Eusebio 61773-270, Brazil; (J.P.V.R.); (V.B.P.G.); (R.N.)
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA;
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | | | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Henrique Douglas Melo Coutinho
- Departament of Biological Chemistry, Universidade Regional do Cariri (URCA), Crato 63105-010, Brazil; (C.D.d.M.O.-T.); (G.G.A.); (G.M.S.); (D.S.A.); (T.F.M.); (S.R.T.); (I.R.A.d.M.)
| | - Teresinha Gonçalves da Silva
- Departamento de Antibióticos, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, Brazil; (J.E.G.S.); (T.G.d.S.)
| |
Collapse
|
64
|
Chauhan M, Singh RP, Sonali, Yadav B, Shekhar S, Kumar L, Mehata AK, Jhawat V, Dutt R, Garg V, Kailashiya V, Muthu MS. Dual-targeted transferrin and AS1411 aptamer conjugated micelles for improved therapeutic efficacy and imaging of brain cancer. Colloids Surf B Biointerfaces 2023; 231:113544. [PMID: 37769388 DOI: 10.1016/j.colsurfb.2023.113544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023]
Abstract
Brain tumors represent an aggressive form of cancer, posing significant challenges in achieving complete remission. Development of advanced therapies is crucial for improving clinical outcomes in cancer patients. This study aimed to create a novel treatment approach using dual-targeted transferrin (TF) and AS1411 conjugated micelles, designed to enhance therapeutic effectiveness of docetaxel (DTX) and facilitate gadolinium (Gd) based imaging in brain cancer. Micelles were prepared using a slightly modified solvent-casting method, and the dual-targeting ligands were attached to the micelle's surface through a physical adsorption process. Average particle size of micelles ranged from 117.49 ± 3.90-170.38 ± 3.39 nm, with a low polydispersity index. Zeta potential ranged from - 1.5 ± 0.02 to - 18.7 ± 0.04 mV. Encapsulation efficiency of DTX in micelles varied from 92.64 ± 4.22-79.77 ± 4.13 %. Simultaneously, encapsulation of Gd in micelles was found to be 48.27 ± 3.18-58.52 ± 3.17, respectively. In-vitro drug release studies showed a biphasic sustained release profile, with DTX and Gd release continuing up to 72 h with their t50 % at 4.95, 11.29, and 24.14 h for GDTP, GDTP-TF and GDTP-TF-AS1411 micelles, respectively. Cytotoxicity effect of GDTP-TF-AS1411 micelles has shown significant improvement (P < 0.001) and reduced IC50 value up to 0.19 ± 0.14 μg/ml compared to Taxotere® (2.73 ± 0.73 μg/ml). Theranostic study revealed higher accumulation of GDTP-TF and GDTP-TF-AS1411 micelles free GD treated animal brains. The AUC of GDTP-TF-AS1411 micelles exhibited 23.79 ± 17.82 μg.h/ml higher than Taxotere® (14.14 ± 10.59 μg.h/ml). These findings direct enhanced effectiveness in brain cancer therapy leading to improved therapeutics in brain cancer patients. The combined targeted ligands and therapeutic agents strategy can direct advancement in brain cancer therapy and offer improved therapy for patients.
Collapse
Affiliation(s)
- Mahima Chauhan
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Rahul Pratap Singh
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India.
| | - Sonali
- Guru Teg Bahadur Hospital, GTB Enclave, Dilshad Garden, New Delhi, Delhi 110095, India
| | - Bhavna Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Saurabh Shekhar
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Lokesh Kumar
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Vikas Jhawat
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Rohit Dutt
- Department of Pharmacy, School of Medical and Allied Sciences, GD Goenka University, Gurugram 122103, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Vikas Kailashiya
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
65
|
Verimli N, Goralı Sİ, Abisoglu B, Altan CL, Sucu BO, Karatas E, Tulek A, Bayraktaroglu C, Beker MC, Erdem SS. Development of light and pH-dual responsive self-quenching theranostic SPION to make EGFR overexpressing micro tumors glow and destroy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 248:112797. [PMID: 37862898 DOI: 10.1016/j.jphotobiol.2023.112797] [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: 06/01/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
Drug resistant and undetectable tumors easily escape treatment leading metastases and/or recurrence of the lethal disease. Therefore, it is vital to diagnose and destroy micro tumors using simple yet novel approaches. Here, we present fluorescence-based detection and light-based destruction of cancer cells that are known to be resistant to standard therapies. We developed a superparamagnetic iron oxide nanoparticle (SPION)-based theranostic agent that is composed of self-quenching light activated photosensitizer (BPD) and EGFR targeting ligand (Anti-EGFR ScFv or GE11 peptide). Photosensitizer (BPD) was immobilized to PEG-PEI modified SPION with acid-labile linker. Prior to stimulation of the theranostic system by light its accumulation within cancer cells is vital since BPD phototoxicity and fluorescence is activated by lysosomal proteolysis. As BPD is cleaved, the system switches from off to on position which triggers imaging and therapy. Targeting, therapeutic and diagnostic features of the theranostic system were evaluated in high and moderate level EGFR expressing pancreatic cancer cell lines. Our results indicate that the system distinguishes high and moderate EGFR expression levels and yields up to 4.3-fold increase in intracellular fluorescence intensity. Amplification of fluorescence signal was as low as 1.3-fold in the moderate or no EGFR expressing cell lines. Anti-EGFR ScFv targeted SPION caused nearly 2-fold higher cell death via apoptosis in high EGFR expressing Panc-1 cell line. The developed system, possessing advanced targeting, enhanced imaging and effective therapeutic features, is a promising candidate for multi-mode detection and destruction of residual drug-resistant cancer cells.
Collapse
Affiliation(s)
- Nihan Verimli
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey
| | - S İrem Goralı
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey
| | - Beyza Abisoglu
- Department of Chemical Engineering, Yeditepe University, Atasehir, Istanbul 34755, Turkey
| | - Cem Levent Altan
- Department of Chemical Engineering, Yeditepe University, Atasehir, Istanbul 34755, Turkey
| | - Bilgesu Onur Sucu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul Medipol University, Istanbul, Turkey; Center of Drug Discovery and Development, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Ersin Karatas
- Ağrı İbrahim Çeçen University, Patnos Vocational School, Department of Medical Services and Techniques, Ağrı, Turkey
| | - Ahmet Tulek
- Iğdır University, Vocational School of Health Services, Department of Care Services, Iğdır, Turkey
| | - Cigdem Bayraktaroglu
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey
| | - Mustafa Caglar Beker
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey
| | - S Sibel Erdem
- Research Institute for Health Science and Technologies (SABITA), 34810 Istanbul, Turkey; International School of Medicine, Medical Biochemistry, Istanbul Medipol University, 34810 Istanbul, Turkey.
| |
Collapse
|
66
|
Abeesh P, Bouvet P, Guruvayoorappan C. AS1411 aptamer tagged PEGylated liposomes as a smart nanocarrier for tumor-specific delivery of Withaferin A for mitigating pulmonary metastasis. BIOMATERIALS ADVANCES 2023; 154:213661. [PMID: 37879185 DOI: 10.1016/j.bioadv.2023.213661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/17/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
Metastasis is the most challenging health problem contributing to about 90 % of cancer-related deaths worldwide. Metastatic tumors are highly aggressive and resistant to the most available therapeutic options. Hence, innovative therapeutic approaches are required to target metastatic tumors selectively. In this study, we prepared AS1411 functionalized Withaferin A loaded PEGylated nanoliposomes (ALW) and investigated its therapeutic effect in B16F10 induced in pulmonary metastasis mice models. The prepared formulations' size and morphological properties were evaluated using dynamic light scattering system and Transmission electron microscope. ALW had spherical-shaped nanosized particles with a size of 118 nm and an encapsulation efficacy of 82.5 %. TEM analysis data indicated that ALW has excellent dispersibility and uniform spherical nano-size particles. ALW inhibited cell viability, and induced cell apoptosis of B16F10. In vivo, the pulmonary metastasis study in C57BL/6 mice revealed that the ALW significantly (p < 0.01) improved the encapsulated WA anti-metastatic activity and survival rate compared to WA or LW treated groups. ALW significantly (p < 0.01) downregulated the levels of IL-6, TNF-α, and IL-1β and significantly reduced the lung collagen hydroxyproline, hexosamine, and uronic acid content in metastatic tumor bearing animals compared to WA or LW. Gene expression levels of MMPs and NF-κB were downregulated in ALW treated metastatic pulmonary tumor-bearing mice. These findings demonstrate that the AS1411 functionalized Withaferin A loaded PEGylated nanoliposomes could be a promising nanoliposomal formulation for targeting metastatic tumors.
Collapse
Affiliation(s)
- Prathapan Abeesh
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College post, Thiruvananthapuram 695011, Kerala, India (Recognized Research Centre, University of Kerala)
| | - Phillipe Bouvet
- Centre de Recherche en Cancerologie de Lyon, Universite de Lyon 1, Inserm U1052, CNRS UMR5286 Centre Leon Berard, CEDEX 08, F-69373 Lyon, France; Ecole Normale Superieur de Lyon, Universite de Lyon 1, F-69007 Lyon, France
| | - Chandrasekaran Guruvayoorappan
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College post, Thiruvananthapuram 695011, Kerala, India (Recognized Research Centre, University of Kerala).
| |
Collapse
|
67
|
Zheng S, Zhang X, Pang Z, Liu J, Liu S, Sheng R. Anti-Pan-Rspo Chimeric Protein-Conjugated Albumin Nanoparticle Provides Promising Opportunities in Cancer Targeted Therapy. Adv Healthc Mater 2023; 12:e2301441. [PMID: 37414582 DOI: 10.1002/adhm.202301441] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Indexed: 07/08/2023]
Abstract
Rspos (R-spondins) belong to a family of secreted proteins that causes various cancers via interacting the corresponding receptors. However, targeted therapeutic approaches against Rspos are largely lacking. In this study, a chimeric protein Rspo-targeting anticancer chimeric protein (RTAC) is originally designed, engineered, and characterized. RTAC shows satisfactory anticancer effects through inhibition of pan-Rspo-mediated Wnt/β-catenin signaling activation both in vitro and in vivo. Furthermore, a conceptually novel antitumor strategy distinct from traditional drug delivery systems that release drugs inside tumor cells is proposed. A special "firewall" nano-system is designed to enrich on tumor cell surface and cover the plasma membrane, rather than undergoing endocytosis, to block oncogenic Rspos from binding to receptors. Cyclic RGD (Arg-Gly-Asp) peptide-linked globular cluster serum albumin nanoparticles (SANP) are integrated as a vehicle for conjugating RTAC (SANP-RTAC/RGD) for tumor tissue targeting. These nanoparticles can adhere to the tumor cell surface and enable RTAC to locally capture free Rspos with high spatial efficiency and selectivity to antagonize cancer progression. Therefore, this approach offers a new nanomedical anticancer route and obtains the "dual-targeting" capability for effective tumor clearance and low potential toxicity. This study presents a proof-of-concept for anti-pan-Rspo therapy and a nanoparticle-integrated paradigm for targeted cancer treatment.
Collapse
Affiliation(s)
- Shaoqin Zheng
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Shenyang, Liaoning, 110819, China
| | - Xi Zhang
- College of Science, Northeastern University, 3-11 Wenhua Road, Shenyang, Liaoning, 110004, China
| | - Zhongqiu Pang
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Shenyang, Liaoning, 110819, China
| | - Jidong Liu
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Shenyang, Liaoning, 110819, China
| | - Siyu Liu
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Shenyang, Liaoning, 110819, China
| | - Ren Sheng
- College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Shenyang, Liaoning, 110819, China
| |
Collapse
|
68
|
Sell M, Lopes AR, Escudeiro M, Esteves B, Monteiro AR, Trindade T, Cruz-Lopes L. Application of Nanoparticles in Cancer Treatment: A Concise Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2887. [PMID: 37947732 PMCID: PMC10650201 DOI: 10.3390/nano13212887] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Timely diagnosis and appropriate antitumoral treatments remain of utmost importance, since cancer remains a leading cause of death worldwide. Within this context, nanotechnology offers specific benefits in terms of cancer therapy by reducing its adverse effects and guiding drugs to selectively target cancer cells. In this comprehensive review, we have summarized the most relevant novel outcomes in the range of 2010-2023, covering the design and application of nanosystems for cancer therapy. We have established the general requirements for nanoparticles to be used in drug delivery and strategies for their uptake in tumor microenvironment and vasculature, including the reticuloendothelial system uptake and surface functionalization with protein corona. After a brief review of the classes of nanovectors, we have covered different classes of nanoparticles used in cancer therapies. First, the advances in the encapsulation of drugs (such as paclitaxel and fisetin) into nanoliposomes and nanoemulsions are described, as well as their relevance in current clinical trials. Then, polymeric nanoparticles are presented, namely the ones comprising poly lactic-co-glycolic acid, polyethylene glycol (and PEG dilemma) and dendrimers. The relevance of quantum dots in bioimaging is also covered, namely the systems with zinc sulfide and indium phosphide. Afterwards, we have reviewed gold nanoparticles (spheres and anisotropic) and their application in plasmon-induced photothermal therapy. The clinical relevance of iron oxide nanoparticles, such as magnetite and maghemite, has been analyzed in different fields, namely for magnetic resonance imaging, immunotherapy, hyperthermia, and drug delivery. Lastly, we have covered the recent advances in the systems using carbon nanomaterials, namely graphene oxide, carbon nanotubes, fullerenes, and carbon dots. Finally, we have compared the strategies of passive and active targeting of nanoparticles and their relevance in cancer theranostics. This review aims to be a (nano)mark on the ongoing journey towards realizing the remarkable potential of different nanoparticles in the realm of cancer therapeutics.
Collapse
Affiliation(s)
- Mariana Sell
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
| | - Ana Rita Lopes
- Faculty of Dental Medicine, Portuguese Catholic University, 3504-505 Viseu, Portugal;
| | - Maria Escudeiro
- Abel Salazar Biomedical Institute, University of Porto, 4050-313 Porto, Portugal;
| | - Bruno Esteves
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| | - Ana R. Monteiro
- Centro de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain;
| | - Tito Trindade
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luísa Cruz-Lopes
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| |
Collapse
|
69
|
Kozlov D, Rodimova S, Kuznetsova D. The Role of MicroRNAs in Liver Functioning: from Biogenesis to Therapeutic Approaches (Review). Sovrem Tekhnologii Med 2023; 15:54-79. [PMID: 39967915 PMCID: PMC11832066 DOI: 10.17691/stm2023.15.5.06] [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: 07/25/2023] [Indexed: 01/03/2025] Open
Abstract
Molecular diagnostics based on small non-coding RNA molecules (in particular microRNA) is a new direction in modern biomedicine and is considered a promising method for identification of a wide range of pathologies at an early stage, clinical phenotype assessment, as well as monitoring the course of the disease, evaluation of therapy efficacy and the risk of the disease recurrence. Currently, the role of microRNAs as the most important epigenetic regulator in cancer development has been proven within the studies of normal and pathogenic processes. However, currently, there are insignificant studies devoted to studying the role of microRNAs in functioning of other organs and tissues, as well as to development of possible therapeutic approaches based on microRNAs. A huge number of metabolic processes in the liver are controlled by microRNAs, which creates enormous potential for the use of microRNAs as a diagnostic marker and makes it a target for therapeutic intervention in metabolic, oncological, and even viral diseases of this organ. This review examines various aspects of biological functions of microRNAs in different types of liver cells. Both canonical and non-canonical pathways of biogenesis, epigenetic regulation mediated by microRNAs, as well as the microRNAs role in intercellular communication and the course of viral diseases are shown. The potential of microRNAs as a diagnostic marker for various liver pathologies is described, as well as therapeutic approaches and medicines based on microRNAs, which are approved for clinical use and currently being developed.
Collapse
Affiliation(s)
- D.S. Kozlov
- Laboratory Assistant, Scientific Laboratory of Molecular Biotechnologies, I Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Student, Institute of Biology and Biomedicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - S.A. Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Junior Researcher, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- PhD, Head of the Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of the Research Laboratory for Molecular Genetic Researches, Institute of Clinical Medicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| |
Collapse
|
70
|
Wang C, Zhang Y. Current Application of Nanoparticle Drug Delivery Systems to the Treatment of Anaplastic Thyroid Carcinomas. Int J Nanomedicine 2023; 18:6037-6058. [PMID: 37904863 PMCID: PMC10613415 DOI: 10.2147/ijn.s429629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/18/2023] [Indexed: 11/01/2023] Open
Abstract
Anaplastic thyroid carcinomas (ATCs) are a rare subtype of thyroid cancers with a low incidence but extremely high invasiveness and fatality. The treatment of ATCs is very challenging, and currently, a comprehensive individualized therapeutic strategy involving surgery, radiotherapy (RT), chemotherapy, BRAF/MEK inhibitors (BRAFi/MEKi) and immunotherapy is preferred. For ATC patients in stage IVA/IVB, a surgery-based comprehensive strategy may provide survival benefits. Unfortunately, ATC patients in IVC stage barely get benefits from the current treatment. Recently, nanoparticle delivery of siRNAs, targeted drugs, cytotoxic drugs, photosensitizers and other agents is considered as a promising anti-cancer treatment. Nanoparticle drug delivery systems have been mainly explored in the treatment of differentiated thyroid cancer (DTC). With the rapid development of drug delivery techniques and nanomaterials, using hybrid nanoparticles as the drug carrier to deliver siRNAs, targeted drugs, immune drugs, chemotherapy drugs and phototherapy drugs to ATC patients have become a hot research field. This review aims to describe latest findings of nanoparticle drug delivery systems in the treatment of ATCs, thus providing references for the further analyses.
Collapse
Affiliation(s)
- Chonggao Wang
- Department of Thyroid Surgery, Nanjing Hospital of Chinese Medicine, Nanjing, 210001, People’s Republic of China
- School of Medicine, Southeast University, Nanjing, 210001, People’s Republic of China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210009, People’s Republic of China
| |
Collapse
|
71
|
Strużyńska L. Dual Implications of Nanosilver-Induced Autophagy: Nanotoxicity and Anti-Cancer Effects. Int J Mol Sci 2023; 24:15386. [PMID: 37895066 PMCID: PMC10607027 DOI: 10.3390/ijms242015386] [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: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
In recent years, efforts have been made to identify new anti-cancer therapies. Various types of nanomaterials, including silver nanoparticles (AgNPs), are being considered as an option. In addition to its well-known antibacterial activity, AgNPs exhibit cytotoxic potential in both physiological and cancer cells by inducing stress-mediated autophagy and apoptotic cell death. A rapidly growing collection of data suggests that the proper regulation of autophagic machinery may provide an efficient tool for suppressing the development of cancer. In this light, AgNPs have emerged as a potential anti-cancer agent to support therapy of the disease. This review summarizes current data indicating the dual role of AgNP-induced autophagy and highlights factors that may influence its protective vs. its toxic potential. It also stresses that our understanding of the cellular and molecular mechanisms of autophagy machinery in cancer cells, as well as AgNP-triggered autophagy in both normal and diseased cells, remains insufficient.
Collapse
Affiliation(s)
- Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str., 02-106 Warsaw, Poland
| |
Collapse
|
72
|
Voltà-Durán E, Alba-Castellón L, Serna N, Casanova I, López-Laguna H, Gallardo A, Sánchez-Chardi A, Villaverde A, Unzueta U, Vázquez E, Mangues R. High-precision targeting and destruction of cancer-associated PDGFR-β + stromal fibroblasts through self-assembling, protein-only nanoparticles. Acta Biomater 2023; 170:543-555. [PMID: 37683965 DOI: 10.1016/j.actbio.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023]
Abstract
The need for more effective and precision medicines for cancer has pushed the exploration of new materials appropriate for drug delivery and imaging, and alternative receptors for targeting. Among the most promising strategies, finding suitable cell surface receptors and targeting agents for cancer-associated platelet derived growth factor receptor β (PDGFR-β)+ stromal fibroblasts is highly appealing. As a neglected target, this cell type mechanically and biologically supports the growth, progression, and infiltration of solid tumors in non-small cell lung, breast, pancreatic, and colorectal cancers. We have developed a family of PDGFR-β-targeted nanoparticles based on biofabricated, self-assembling proteins, upon hierarchical and iterative selective processes starting from four initial candidates. The modular protein PDGFD-GFP-H6 is well produced in recombinant bacteria, resulting in structurally robust oligomeric particles that selectively penetrates into PDGFR-β+ stromal fibroblasts in a dose-dependent manner, by means of the PDGFR-β ligand PDGFD. Upon in vivo administration, these GFP-carrying protein nanoparticles precisely accumulate in tumor tissues and enlighten them for IVIS observation. When GFP is replaced by a microbial toxin, selective tumor tissue destruction is observed associated with a significant reduction in tumor volume growth. The presented data validate the PDGFR-β/PDGFD pair as a promising toolbox for targeted drug delivery in the tumor microenvironment and oligomeric protein nanoparticles as a powerful instrument to mediate highly selective biosafe targeting in cancer through non-cancer cells. STATEMENT OF SIGNIFICANCE: We have developed a transversal platform for nanoparticle-based drug delivery into cancer-associated fibroblasts. This is based on the engineered modular protein PDGFD-GFP-H6 that spontaneously self-assemble and selectively penetrates into PDGFR-β+ stromal fibroblasts in a dose-dependent manner, by means of the PDGFR-β ligand PDGFD. In vivo, these protein nanoparticles accumulate in tumor and when incorporating a microbial toxin, they destroy tumor tissues with a significant reduction in tumor volume, in absence of side toxicities. The data presented here validate the PDGFR-β/PDGFD pair as a fully versatile toolbox for targeted drug delivery in the tumor microenvironment intended as a synergistic treatment.
Collapse
Affiliation(s)
- Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Lorena Alba-Castellón
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona 08041, Spain; Josep Carreras Leukaemia Research Institute, Barcelona 08025, Spain.
| | - Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Isolda Casanova
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona 08041, Spain; Josep Carreras Leukaemia Research Institute, Barcelona 08025, Spain
| | - Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Alberto Gallardo
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona 08041, Spain; Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
| | - Alejandro Sánchez-Chardi
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona 08028, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona 08041, Spain; Josep Carreras Leukaemia Research Institute, Barcelona 08025, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain; Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona 08041, Spain; Josep Carreras Leukaemia Research Institute, Barcelona 08025, Spain.
| |
Collapse
|
73
|
Kozak A, Mikhaylov G, Khodakivskyi P, Goun E, Turk B, Vasiljeva O. A New Cathepsin D Targeting Drug Delivery System Based on Immunoliposomes Functionalized with Lipidated Pepstatin A. Pharmaceutics 2023; 15:2464. [PMID: 37896224 PMCID: PMC10609775 DOI: 10.3390/pharmaceutics15102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Cathepsin D is an aspartic protease and one of the most abundant proteases. It is overexpressed in many cancers and plays an important role in tumor development, progression, and metastasis. While it is a physiologically intracellular protein, cathepsin D is secreted into the extracellular matrix under pathological conditions, making it an appealing target for drug delivery systems. Here, we present the development and evaluation of a new delivery system for tumor targeting based on immunoliposomes functionalized with pepstatin A-a natural peptide inhibitor of cathepsin D. A lipid tail was added to pepstatin A, enabling its incorporation into the liposomal lipid bilayer. The successful targeting of cathepsin D was confirmed using recombinant cathepsin D and in tumor cell lines, showing the feasibility of this targeting approach and its potential for in vivo use in theragnostic applications.
Collapse
Affiliation(s)
- Andreja Kozak
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia
- SwissLumix SARL, 1015 Lausanne, Switzerland
| | - Pavlo Khodakivskyi
- Department of Chemistry, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Elena Goun
- SwissLumix SARL, 1015 Lausanne, Switzerland
- Department of Chemistry, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, 1000 Ljubljana, Slovenia
| |
Collapse
|
74
|
Lebreton V, Legeay S, Vasylaki A, Lagarce F, Saulnier P. Protein corona formation on lipidic nanocapsules: Influence of the interfacial PEG repartition. Eur J Pharm Sci 2023; 189:106537. [PMID: 37490974 DOI: 10.1016/j.ejps.2023.106537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/04/2023] [Accepted: 07/22/2023] [Indexed: 07/27/2023]
Abstract
The parameters currently used for characterization of nanoparticles, such as size and zeta potential, were not able to reflect the performance of a nanocarrier in the biological environment. Therefore, more thorough in vitro characterization is required to predict their behavior in vivo, where nanoparticles acquire a new biological identity due to interactions with biomolecules. In this present study, we performed in vitro characterization in biological fluids for lipid nanocapsules (LNCs) with varying means sizes (50 nm and 100 nm), different electrical surface charges and different Poly Ethylene Glycol (PEG) compositions. Then, different methods were applied to show the impact of the protein corona formation on LNCs. Even if all formulations attached to plasmatic proteins, a higher thickness of corona and highest protein binding was observed for certain LNC50 formulations. A better knowledge of the phenomenon of protein adsorption over NPs in the plasmatic media is a cornerstone of clinical translation. In fact, after short blood circulation time, it is not the initially designed nanoparticle but the complex nanoparticle bearing its protein corona which circulates to reach its target.
Collapse
Affiliation(s)
- Vincent Lebreton
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France.
| | - Samuel Legeay
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France
| | | | - Fredéric Lagarce
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France
| | - Patrick Saulnier
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France
| |
Collapse
|
75
|
Rathnayake K, Patel U, Hunt EC, Singh N. Fabrication of a Dual-Targeted Liposome-Coated Mesoporous Silica Core-Shell Nanoassembly for Targeted Cancer Therapy. ACS OMEGA 2023; 8:34481-34498. [PMID: 37779923 PMCID: PMC10536893 DOI: 10.1021/acsomega.3c02901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Nanoparticles have been suggested as drug-delivery systems for chemotherapeutic drugs to allow for controlled drug release profiles and selectivity to target cancer cells. In addition, nanoparticles can be used for the in situ generation and amplification of reactive oxygen species (ROS), which have been shown to be a promising strategy for cancer treatment. Thus, a targeted nanoscale drug-delivery platform could be used to synergistically improve cancer treatment by the action of chemotherapeutic drugs and ROS generation. Herein, we propose a promising chemotherapy strategy where the drug-loaded nanoparticles generate high doses of ROS together with the loaded ROS-generating chemotherapeutic drugs, which can damage the mitochondria and activate cell death, potentiating the therapeutic outcome in cancer therapy. In the present study, we have developed a dual-targeted drug-delivery nanoassembly consisting of a mesoporous silica core loaded with the chemotherapeutic, ROS-generating drug, paclitaxel (Px), and coated with a liposome layer for controlled drug release. Two different lung cancer-targeting ligands, folic acid and peptide GE11, were used to target the overexpressed nonsmall lung cancer receptors to create the final nanoassembly (MSN@Px) L-GF. Upon endocytosis by the cancer cells, the liposome layer was degraded by the intracellular lipases, and the drug was rapidly released at a rate of 65% within the first 20 h. In vitro studies confirmed that this nanoassembly was 8-fold more effective in cancer therapy compared to the free drug Px.
Collapse
Affiliation(s)
- Kavini Rathnayake
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Unnati Patel
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Emily C. Hunt
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Nirupama Singh
- Department of Chemistry, The
University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| |
Collapse
|
76
|
Bouquerel C, Dubrova A, Hofer I, Phan DTT, Bernheim M, Ladaigue S, Cavaniol C, Maddalo D, Cabel L, Mechta-Grigoriou F, Wilhelm C, Zalcman G, Parrini MC, Descroix S. Bridging the gap between tumor-on-chip and clinics: a systematic review of 15 years of studies. LAB ON A CHIP 2023; 23:3906-3935. [PMID: 37592893 DOI: 10.1039/d3lc00531c] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.
Collapse
Affiliation(s)
- Charlotte Bouquerel
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Anastasiia Dubrova
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Isabella Hofer
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Duc T T Phan
- Biomedicine Design, Pfizer Inc., San Diego, CA, USA
| | - Moencopi Bernheim
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Ségolène Ladaigue
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Charles Cavaniol
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Luc Cabel
- Institut Curie, Department of Medical Oncology, 26 rue d'Ulm, 75005, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Claire Wilhelm
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Gérard Zalcman
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Université Paris Cité, Thoracic Oncology Department, INSERM CIC1425, Bichat Hospital, Cancer Institute AP-HP. Nord, Paris, France.
| | - Maria Carla Parrini
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Stéphanie Descroix
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| |
Collapse
|
77
|
El Moukhtari SH, Garbayo E, Amundarain A, Pascual-Gil S, Carrasco-León A, Prosper F, Agirre X, Blanco-Prieto MJ. Lipid nanoparticles for siRNA delivery in cancer treatment. J Control Release 2023; 361:130-146. [PMID: 37532145 DOI: 10.1016/j.jconrel.2023.07.054] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/08/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
RNA-based therapies, and siRNAs in particular, have attractive therapeutic potential for cancer treatment due to their ability to silence genes that are imperative for tumor progression. To be effective and solve issues related to their poor half-life and poor pharmacokinetic properties, siRNAs require adequate drug delivery systems that protect them from degradation and allow intracellular delivery. Among the various delivery vehicles available, lipid nanoparticles have emerged as the leading choice. These nanoparticles consist of cholesterol, phospholipids, PEG-lipids and most importantly ionizable cationic lipids. These ionizable lipids enable the binding of negatively charged siRNA, resulting in the formation of stable and neutral lipid nanoparticles with exceptionally high encapsulation efficiency. Lipid nanoparticles have demonstrated their effectiveness and versatility in delivering not only siRNAs but also multiple RNA molecules, contributing to their remarkable success. Furthermore, the advancement of efficient manufacturing techniques such as microfluidics, enables the rapid mixing of two miscible solvents without the need for shear forces. This facilitates the reproducible production of lipid nanoparticles and holds enormous potential for scalability. This is shown by the increasing number of preclinical and clinical trials evaluating the potential use of siRNA-LNPs for the treatment of solid and hematological tumors as well as in cancer immunotherapy. In this review, we provide an overview of the progress made on siRNA-LNP development for cancer treatment and outline the current preclinical and clinical landscape in this area. Finally, the translational challenges required to bring siRNA-LNPs further into the clinic are also discussed.
Collapse
Affiliation(s)
- Souhaila H El Moukhtari
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Ane Amundarain
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Simón Pascual-Gil
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Arantxa Carrasco-León
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Felipe Prosper
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain; Departmento de Hematología and CCUN, Clínica Universidad de Navarra, University of Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
| | - Xabier Agirre
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
78
|
Zhao DK, Liang J, Huang XY, Shen S, Wang J. Organoids technology for advancing the clinical translation of cancer nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1892. [PMID: 37088100 DOI: 10.1002/wnan.1892] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/25/2023]
Abstract
The past decades have witnessed the rapid development and widespread application of nanomedicines in cancer treatment; however, the clinical translation of experimental findings has been low, as evidenced by the low percentage of commercialized nanomedicines. Incomplete understanding of nanomedicine-tumor interactions and inappropriate evaluation models are two important challenges limiting the clinical translation of cancer nanomedicines. Currently, nanomedicine-tumor interaction and therapeutic effects are mainly investigated using cell lines or mouse models, which do not recapitulate the complex tumor microenvironment in human patients. Thus, information obtained from cell lines and mouse models cannot provide adequate guidance for the rational redesign of nanomedicine. Compared with other preclinical models, tumor organoids constructed from patient-derived tumor tissues are superior in retaining the key histopathological, genetic, and phenotypic features of the parent tumor. We speculate that organoid technology would help elucidate nanomedicine-tumor interaction in the tumor microenvironment and guide the design of nanomedicine, making it a reliable tool to accurately predict drug responses in patients with cancer. This review highlighted the advantages of drug delivery systems in cancer treatment, challenges limiting the clinical translation of antitumor nanomedicines, and potential application of patient-derived organoids (PDO) in nanomedicine. We propose that combining organoids and nanotechnology would facilitate the development of safe and effective cancer nanomedicines and accelerate their clinical application. This review discussed the potential translational value of integrative research using organoids and cancer nanomedicine. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Jie Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiao-Yi Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| |
Collapse
|
79
|
Song S, Sun D, Wang H, Wang J, Yan H, Zhao X, Fawcett JP, Xu X, Cai D, Gu J. Full-profile pharmacokinetics, anticancer activity and toxicity of an extended release trivalent PEGylated irinotecan prodrug. Acta Pharm Sin B 2023; 13:3444-3453. [PMID: 37655324 PMCID: PMC10466002 DOI: 10.1016/j.apsb.2023.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/22/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Irinotecan is an anticancer topoisomerase I inhibitor that acts as a prodrug of the active metabolite, SN-38. Unfortunately, the limited utility of irinotecan is attributed to its pH-dependent stability, short half-life and dose-limiting toxicity. To address this problem, a novel trivalent PEGylated prodrug (PEG-[Irinotecan]3) has been synthesized and its full-profile pharmacokinetics, antitumor activity and toxicity compared with those of irinotecan. The results show that after intravenous administration to rats, PEG-[Irinotecan]3 undergoes stepwise loss of irinotecan to form PEG-[Irinotecan]3‒x (x = 1,2) and PEG-[linker] during which time the released irinotecan undergoes conversion to SN-38. As compared with conventional irinotecan, PEG-[Irinotecan]3 displays extended release of irinotecan and efficient formation of SN-38 with significantly improved AUC and half-life. In a colorectal cancer-bearing model in nude mice, the tumor concentrations of irinotecan and SN-38 produced by PEG-[Irinotecan]3 were respectively 86.2 and 2293 times higher at 48 h than produced by irinotecan. In summary, PEG-[Irinotecan]3 displays superior pharmacokinetic characteristics and antitumor activity with lower toxicity than irinotecan. This supports the view that PEG-[Irinotecan]3 is a superior anticancer drug to irinotecan and it has entered the phase II trial stage.
Collapse
Affiliation(s)
- Shiwen Song
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Dong Sun
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Hong Wang
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- Center for Food and Drug Inspection of NMPA, Changchun 130012, China
| | | | - Huijing Yan
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - Xuan Zhao
- JenKem Technology Co., Ltd., Tianjin 300450, China
| | - John Paul Fawcett
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Xu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Deqi Cai
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
- Beijing Institute of Drug Metabolism, Beijing 102209, China
| |
Collapse
|
80
|
Stahl P, Kollenda S, Sager J, Schmidt L, Schroer MA, Stauber RH, Epple M, Knauer SK. Tuning Nanobodies' Bioactivity: Coupling to Ultrasmall Gold Nanoparticles Allows the Intracellular Interference with Survivin. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300871. [PMID: 37035950 DOI: 10.1002/smll.202300871] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Indexed: 06/19/2023]
Abstract
Nanobodies are highly affine binders, often used to track disease-relevant proteins inside cells. However, they often fail to interfere with pathobiological functions, required for their clinical exploitation. Here, a nanobody targeting the disease-relevant apoptosis inhibitor and mitosis regulator Survivin (SuN) is utilized. Survivin's multifaceted functions are regulated by an interplay of dynamic cellular localization, dimerization, and protein-protein interactions. However, as Survivin harbors no classical "druggable" binding pocket, one must aim at blocking extended protein surface areas. Comprehensive experimental evidence demonstrates that intracellular expression of SuN allows to track Survivin at low nanomolar concentrations but failed to inhibit its biological functions. Small angle X-ray scattering of the Survivin-SuN complex locates the proposed interaction interface between the C-terminus and the globular domain, as such not blocking any pivotal interaction. By clicking multiple SuN to ultrasmall (2 nm) gold nanoparticles (SuN-N), not only intracellular uptake is enabled, but additionally, Survivin crosslinking and interference with mitotic progression in living cells are also enabled. In sum, it is demonstrated that coupling of nanobodies to nanosized scaffolds can be universally applicable to improve their function and therapeutic applicability.
Collapse
Affiliation(s)
- Paul Stahl
- Molecular Biology II, Department of Biology, Center of Medical Biotechnology (ZMB), University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| | - Sebastian Kollenda
- Inorganic Chemistry, Department of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45141, Essen, Germany
| | - Jonas Sager
- Inorganic Chemistry, Department of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45141, Essen, Germany
| | - Laura Schmidt
- Molecular Biology II, Department of Biology, Center of Medical Biotechnology (ZMB), University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| | - Martin A Schroer
- Nanoparticle Process Technology, Department of Engineering, University of Duisburg-Essen, Lotharstr. 1, 47057, Duisburg, Germany
| | - Roland H Stauber
- Molecular and Cellular Oncology/ENT, University Medical Center Mainz (UMM), Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Matthias Epple
- Inorganic Chemistry, Department of Chemistry, Center for Nanointegration Duisburg-Essen (CENIDE) and Center of Medical Biotechnology (ZMB), University of Duisburg-Essen, Universitätsstrasse 7, 45141, Essen, Germany
| | - Shirley K Knauer
- Molecular Biology II, Department of Biology, Center of Medical Biotechnology (ZMB) and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstrasse 5, 45141, Essen, Germany
| |
Collapse
|
81
|
Shen J, Chen G, Zhao L, Huang G, Liu H, Liu B, Miao Y, Li Y. Recent Advances in Nanoplatform Construction Strategy for Alleviating Tumor Hypoxia. Adv Healthc Mater 2023; 12:e2300089. [PMID: 37055912 DOI: 10.1002/adhm.202300089] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Hypoxia is a typical feature of most solid tumors and has important effects on tumor cells' proliferation, invasion, and metastasis. This is the key factor that leads to poor efficacy of different kinds of therapy including chemotherapy, radiotherapy, photodynamic therapy, etc. In recent years, the construction of hypoxia-relieving functional nanoplatforms through nanotechnology has become a new strategy to reverse the current situation of tumor microenvironment hypoxia and improve the effectiveness of tumor treatment. Here, the main strategies and recent progress in constructing nanoplatforms are focused on to directly carry oxygen, generate oxygen in situ, inhibit mitochondrial respiration, and enhance blood perfusion to alleviate tumor hypoxia. The advantages and disadvantages of these nanoplatforms are compared. Meanwhile, nanoplatforms based on organic and inorganic substances are also summarized and classified. Through the comprehensive overview, it is hoped that the summary of these nanoplatforms for alleviating hypoxia could provide new enlightenment and prospects for the construction of nanomaterials in this field.
Collapse
Affiliation(s)
- Jing Shen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guobo Chen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Linghao Zhao
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Hui Liu
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
82
|
Ghoochani SH, Hosseini HA, Sabouri Z, Soheilifar MH, Neghab HK, Hashemzadeh A, Velayati M, Darroudi M. Zn(II) porphyrin-encapsulated MIL-101 for photodynamic therapy of breast cancer cells. Lasers Med Sci 2023; 38:151. [PMID: 37378703 DOI: 10.1007/s10103-023-03813-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2023] [Indexed: 06/29/2023]
Abstract
The photodynamic treatment is a non-aggressive and clinically accepted procedure for removing selected cancer cells with the activation of a photosensitizer agent at a specific light. In this study, the zinc porphyrin (Zn[TPP]) was prepared and encapsulated into the MIL-101 (Zn[TPP]@MIL-101). It was used in photodynamic therapy (PDT) against MCF-7 breast cancer cells under a red light-emitting diode. The structure, morphology, surface area, and compositional changes were investigated using conventional characterization methods including FTIR, FESEM, EDX, and BET analyses. The MTT assay was performed under light and dark conditions to explore the ability of Zn[TPP]@MIL-101 in PDT. The results have demonstrated the IC50 of 14.3 and 81.6 mg/mL for light and dark groups, respectively. As the IC50 revealed, the Zn[TPP]@MIL-101 could efficiently eradicate cancer cells using PDT.
Collapse
Affiliation(s)
| | | | - Zahra Sabouri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Alireza Hashemzadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahin Velayati
- Chemistry Department, Payame Noor University, Tehran, 19395-4697, Iran
| | - Majid Darroudi
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
83
|
Martínez-Torró C, Alba-Castellón L, Carrasco-Díaz LM, Serna N, Imedio L, Gallardo A, Casanova I, Unzueta U, Vázquez E, Mangues R, Villaverde A. Lymphocyte infiltration and antitumoral effect promoted by cytotoxic inflammatory proteins formulated as self-assembling, protein-only nanoparticles. Biomed Pharmacother 2023; 164:114976. [PMID: 37276641 DOI: 10.1016/j.biopha.2023.114976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/07/2023] Open
Abstract
Two human proteins involved in the inflammatory cell death, namely Gasdermin D (GSDMD) and the Mixed Lineage Kinase Domain-Like (MLKL) protein have been engineered to accommodate an efficient ligand of the tumoral cell marker CXCR4, and a set of additional peptide agents that allow their spontaneous self-assembling. Upon production in bacterial cells and further purification, both proteins organized as stable nanoparticles of 46 and 54 nm respectively, that show, in this form, a moderate but dose-dependent cytotoxicity in cell culture. In vivo, and when administered in mouse models of colorectal cancer through repeated doses, the nanoscale forms of tumor-targeted GSDMD and, at a lesser extent, of MLKL promoted CD8+ and CD20+ lymphocyte infiltration in the tumor and an important reduction of tumor size, in absence of systemic toxicity. The potential of these novel pharmacological agents as anticancer drugs is discussed in the context of synergistic approaches to more effective cancer treatments.
Collapse
Affiliation(s)
- Carlos Martínez-Torró
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Lorena Alba-Castellón
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Luis Miguel Carrasco-Díaz
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Laura Imedio
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Alberto Gallardo
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Department of Pathology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Isolda Casanova
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain.
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| |
Collapse
|
84
|
Zeng S, Tang Q, Xiao M, Tong X, Yang T, Yin D, Lei L, Li S. Cell membrane-coated nanomaterials for cancer therapy. Mater Today Bio 2023; 20:100633. [PMID: 37128288 PMCID: PMC10148189 DOI: 10.1016/j.mtbio.2023.100633] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/01/2023] [Accepted: 04/09/2023] [Indexed: 05/03/2023] Open
Abstract
With the development of nanotechnology, nanoparticles have emerged as a delivery carrier for tumor drug therapy, which can improve the therapeutic effect by increasing the stability and solubility and prolonging the half-life of drugs. However, nanoparticles are foreign substances for humans, are easily cleared by the immune system, are less targeted to tumors, and may even be toxic to the body. As a natural biological material, cell membranes have unique biological properties, such as good biocompatibility, strong targeting ability, the ability to evade immune surveillance, and high drug-carrying capacity. In this article, we review cell membrane-coated nanoparticles (CMNPs) and their applications to tumor therapy. First, we briefly describe CMNP characteristics and applications. Second, we present the characteristics and advantages of different cell membranes as well as nanoparticles, provide a brief description of the process of CMNPs, discuss the current status of their application to tumor therapy, summarize their shortcomings for use in cancer therapy, and propose future research directions. This review summarizes the research progress on CMNPs in cancer therapy in recent years and assesses remaining problems, providing scholars with new ideas for future research on CMNPs in tumor therapy.
Collapse
Affiliation(s)
- Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xinying Tong
- Department of Hemodialysis, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Tao Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Danhui Yin
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
85
|
Tuli HS, Joshi R, Kaur G, Garg VK, Sak K, Varol M, Kaur J, Alharbi SA, Alahmadi TA, Aggarwal D, Dhama K, Jaswal VS, Mittal S, Sethi G. Metal nanoparticles in cancer: from synthesis and metabolism to cellular interactions. JOURNAL OF NANOSTRUCTURE IN CHEMISTRY 2023; 13:321-348. [DOI: 10.1007/s40097-022-00504-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/23/2022] [Indexed: 07/28/2024]
|
86
|
Paradossi G, Grossman R, Riccitelli F, Todaro F, Ram Z, Schioppa S, Domenici F. Toward a theranostic device for gliomas. Biochem Biophys Res Commun 2023; 671:124-131. [PMID: 37300942 DOI: 10.1016/j.bbrc.2023.05.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND In the surgical management of glioblastoma, a highly aggressive and incurable type of brain cancer, identification and treatment of residual tissue is the most common site of disease recurrence. Monitoring and localized treatment are achieved with engineered microbubbles (MBs) by combining ultrasound and fluorescence imaging with actively targeted temozolomide (TMZ) delivery. METHODS The MBs were conjugated with a near-infrared fluorescence probe CF790, cyclic pentapeptide bearing the RGD sequence and a carboxyl-temozolomide, TMZA. The efficiency of adhesion to HUVEC cells was assessed in vitro in realistic physiological conditions of shear rate and vascular dimensions. Cytotoxicity of TMZA-loaded MBs on U87 MG cells and IC50 were assessed by MTT tests. RESULTS We report on the design of injectable poly(vinyl alcohol) echogenic MBs designed as a platform with active targeting ability to tumor tissues, by tethering on the surface a ligand having the tripeptide sequence, RGD. The biorecognition of RGD-MBs onto HUVEC cells is quantitatively proved. Efficient NIR emission from the CF790-decorated MBs was successfully detected. The conjugation on the MBs surface of a specific drug as TMZ is achieved. The pharmacological activity of the coupled-to-surface drug is preserved by controlling the reaction conditions. CONCLUSIONS We present an improved formulation of PVA-MBs to achieve a multifunctional device with adhesion ability, cytotoxicity on glioblastoma cells and supporting imaging.
Collapse
Affiliation(s)
- Gaio Paradossi
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133, Rome, Italy.
| | - Rachel Grossman
- Department of Neurosurgery, Tel Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Francesco Riccitelli
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Federica Todaro
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sara Schioppa
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Fabio Domenici
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", 00133, Rome, Italy
| |
Collapse
|
87
|
Li Y, Zou H, Zheng Z, Liu Z, Hu H, Wu W, Wang T. Advances in the Study of Bioactive Nanoparticles for the Treatment of HCC and Its Postoperative Residual Cancer. Int J Nanomedicine 2023; 18:2721-2735. [PMID: 37250475 PMCID: PMC10216871 DOI: 10.2147/ijn.s399146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Primary hepatocellular carcinoma (HCC, hepatocellular carcinoma) is the third leading cause of tumor death in the world and the second leading cause in China. The high recurrence rate at 5 years after surgery also seriously affects the long-term survival of HCC patients. For reasons such as poor liver function, large tumors, or vascular invasion, only relatively limited palliative treatment is available. Therefore, effective diagnostic and therapeutic strategies are needed to improve the complex microenvironment and block the mechanism of tumor development in order to treat the tumor and prevent recurrence. A variety of bioactive nanoparticles have been shown to have therapeutic effects on hepatocellular carcinoma and have the advantages of improving drug solubility, reducing drug side effects, preventing degradation in the blood, increasing drug exposure time, and reducing drug resistance. The development of bioactive nanoparticles is expected to complete the current clinical therapeutic approach. In this review, we discuss the therapeutic advances of different nanoparticles for hepatocellular carcinoma and discuss their potential for postoperative applications with respect to possible mechanisms of hepatocellular carcinoma recurrence. We further discuss the limitations regarding the application of NPs and the safety of NPs.
Collapse
Affiliation(s)
- Yanxu Li
- Medical College of Yangzhou University, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Hao Zou
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Zekun Zheng
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Zhuoheng Liu
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Huiyuan Hu
- Dalian Medical University, Affiliated Hospital of Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Wei Wu
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou City, Jiangsu Province, People’s Republic of China
| |
Collapse
|
88
|
Mohajer F, Mirhosseini-Eshkevari B, Ahmadi S, Ghasemzadeh MA, Mohammadi Ziarani G, Badiei A, Farshidfar N, Varma RS, Rabiee N, Iravani S. Advanced Nanosystems for Cancer Therapeutics: A Review. ACS APPLIED NANO MATERIALS 2023; 6:7123-7149. [DOI: 10.1021/acsanm.3c00859] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Affiliation(s)
- Fatemeh Mohajer
- Department of Organic Chemistry, Faculty of Chemistry, Alzahra University, Tehran 19938-93973, Iran
| | | | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | | | - Ghodsi Mohammadi Ziarani
- Department of Organic Chemistry, Faculty of Chemistry, Alzahra University, Tehran 19938-93973, Iran
| | - Alireza Badiei
- School of Chemistry, College of Science, University of Tehran, Tehran 14179-35840, Iran
| | - Nima Farshidfar
- Orthodontic Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Rajender S. Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), 1402/2, Liberec 1 461 17, Czech Republic
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia 6150, Australia
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| |
Collapse
|
89
|
Yang T, Zhang X, Yang X, Li Y, Xiang J, Xiang C, Liu Z, Hai L, Huang S, Zhou L, Liang R, Gong P. A mitochondria-targeting self-assembled carrier-free lonidamine nanodrug for redox-activated drug release to enhance cancer chemotherapy. J Mater Chem B 2023; 11:3951-3957. [PMID: 37067569 DOI: 10.1039/d2tb02728c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Mitochondria play a vital role in maintaining cellular homeostasis. In recent years, studies have found that mitochondria have an important role in the occurrence and development of tumors, and targeting mitochondria has become a new strategy for tumor treatment. Lonidamine (LND), as a hexokinase inhibitor, can block the energy supply and destroy mitochondria. However, poor water solubility and low mitochondrial selectivity limit its clinical application. To overcome these obstacles, we report redox-activated self-assembled carrier-free nanoparticles (Cy-TK-LND NPs) based on a small molecule prodrug, in which photosensitizer IR780 (Cy) which targets mitochondria is conjugated to LND via a sensitive thioketal (TK) linker. Intracellular oxidative stress induced by laser radiation leads to the responsive cleavage of Cy-TK-LND NPs, facilitating the release of free LND into mitochondria. Subsequently, LND damages mitochondria, triggering the apoptosis pathway. The results show the effective killing effect of Cy-TK-LND NPs on cancer cells in vitro and in vivo. The IC50 value of irradiated Cy-TK-LND NPs is 5-fold lower than that of free LND. Moreover, tumor tissue section staining results demonstrate that irradiated Cy-TK-LND NPs induce necrosis and apoptosis of tumor cells, upregulate cytochrome C and pro-apoptotic Bax, and downregulate anti-apoptotic Bcl-2. Generally, Cy-TK-LND NPs exhibit efficient mitochondria-targeted delivery to improve the medicinal availability of LND. Accordingly, such a carrier-free prodrug-based nanomedicine holds promise as an effective cancer chemotherapy strategy.
Collapse
Affiliation(s)
- Ting Yang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xianfen Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
- School of Chemical Engineering, Northwest University, Xi'an, 710069, P. R. China.
| | - Xing Yang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Ying Li
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Jingjing Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Chunbai Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Zhongke Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
- Nano Science and Technology Institute, University of Science & Technology of China, Suzhou, 215123, P. R. China
| | - Luo Hai
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, P. R. China
| | - Saipeng Huang
- School of Chemical Engineering, Northwest University, Xi'an, 710069, P. R. China.
| | - Lihua Zhou
- School of Applied Biology, Shenzhen Institute of Technology, No. 1 Jiangjunmao, Shenzhen, 518116, P. R. China.
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China.
| |
Collapse
|
90
|
Kumar AA, Vine KL, Ranson M. Recent Advances in Targeting the Urokinase Plasminogen Activator with Nanotherapeutics. Mol Pharm 2023. [PMID: 37119285 DOI: 10.1021/acs.molpharmaceut.3c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The aberrant proteolytic landscape of the tumor microenvironment is a key contributor of cancer progression. Overexpression of urokinase plasminogen activator (uPA) and/or its associated cell-surface receptor (uPAR) in tumor versus normal tissue is significantly associated with worse clinicopathological features and poorer patient survival across multiple cancer types. This is linked to mechanisms that facilitate tumor cell invasion and migration, via direct and downstream activation of various proteolytic processes that degrade the extracellular matrix─ultimately leading to metastasis. Targeting uPA has thus long been considered an attractive anticancer strategy. However, poor bioavailability of several uPA-selective small-molecule inhibitors has limited early clinical progress. Nanodelivery systems have emerged as an exciting method to enhance the pharmacokinetic (PK) profile of existing chemotherapeutics, allowing increased circulation time, improved bioavailability, and targeted delivery to tumor tissue. Combining uPA inhibitors with nanoparticle-based delivery systems thus offers a remarkable opportunity to overcome existing PK challenges associated with conventional uPA inhibitors, while leveraging potent candidates into novel targeted nanotherapeutics for an improved anticancer response in uPA positive tumors.
Collapse
Affiliation(s)
- Ashna A Kumar
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kara L Vine
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
91
|
Tramontano C, De Stefano L, Rea I. Diatom-Based Nanomedicine for Colorectal Cancer Treatment: New Approaches for Old Challenges. Mar Drugs 2023; 21:md21050266. [PMID: 37233460 DOI: 10.3390/md21050266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Colorectal cancer is among the most prevalent and lethal cancers globally. To address this emergency, countries have developed diffuse screening programs and innovative surgical techniques with a consequent decrease in mortality rates in non-metastatic patients. However, five years after diagnosis, metastatic CRC is still characterized by less than 20% survival. Most patients with metastatic CRC cannot be surgically treated. For them, the only option is treatment with conventional chemotherapies, which cause harmful side effects in normal tissues. In this context, nanomedicine can help traditional medicine overcome its limits. Diatomite nanoparticles (DNPs) are innovative nano-based drug delivery systems derived from the powder of diatom shells. Diatomite is a porous biosilica largely found in many areas of the world and approved by the Food and Drug Administration (FDA) for pharmaceutical and animal feed formulations. Diatomite nanoparticles with a size between 300 and 400 nm were shown to be biocompatible nanocarriers capable of delivering chemotherapeutic agents against specific targets while reducing off-target effects. This review discusses the treatment of colorectal cancer with conventional methods, highlighting the drawbacks of standard medicine and exploring innovative options based on the use of diatomite-based drug delivery systems. Three targeted treatments are considered: anti-angiogenetic drugs, antimetastatic drugs, and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Chiara Tramontano
- Institute of Applied Science and Intelligent Systems (ISASI), National Research Council of Italy-Naples Unit, Via Pietro Castellino 111, 80131 Naples, Italy
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Luca De Stefano
- Institute of Applied Science and Intelligent Systems (ISASI), National Research Council of Italy-Naples Unit, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Ilaria Rea
- Institute of Applied Science and Intelligent Systems (ISASI), National Research Council of Italy-Naples Unit, Via Pietro Castellino 111, 80131 Naples, Italy
| |
Collapse
|
92
|
Sonzini S, Caputo F, Mehn D, Calzolai L, Even Borgos S, Hyldbakk A, Treacher K, Li W, Jackman M, Mahmoudi N, Jayne Lawrence M, Patterson C, Owen D, Ashford M, Akhtar N. In depth characterization of physicochemical critical quality attributes of a clinical drug-dendrimer conjugate. Int J Pharm 2023; 637:122905. [PMID: 37003312 PMCID: PMC10157317 DOI: 10.1016/j.ijpharm.2023.122905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/19/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
A deep and detailed understanding of drug-dendrimer conjugates key properties is needed to define the critical quality attributes that affect drug product performance. The characterization must be executed both in the formulation media and in biological matrices. This, nevertheless, is challenging on account of a very limited number of suitable, established methods for characterizing the physicochemical properties, stability, and interaction with biological environment of complex drug-dendrimer conjugates. In order to fully characterize AZD0466, a drug-dendrimer conjugate currently under clinical development by AstraZeneca, a collaboration was initiated with the European Nanomedicine Characterisation Laboratory to deploy a state-of-the-art multi-step approach to measure physicochemical properties. An incremental complexity characterization approach was applied to two batches of AZD0466 and the corresponding dendrimer not carrying any drug, SPL-8984. Thus, the aim of this work is to guide in depth characterization efforts in the analysis of drug-dendrimer conjugates. Additionally, it serves to highlight the importance of using the adequate complementary techniques to measure physical and chemical stability in both simple and biological media, to drive a complex drug-dendrimer conjugate product from discovery to clinical development.
Collapse
Affiliation(s)
- Silvia Sonzini
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Fanny Caputo
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Univ. Grenoble Alpes, CEA, LETI, F-38000 Grenoble, France
| | - Dora Mehn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Luigi Calzolai
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Sven Even Borgos
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Kevin Treacher
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Weimin Li
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Mark Jackman
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Najet Mahmoudi
- Rutherford Appleton Laboratory, ISIS Facility, Science and Technology Facilities Council, Didcot OX11 0QX, UK
| | - M Jayne Lawrence
- Division of Pharmacy & Optometry and the North West Centre for Advanced Drug Delivery (NoWCADD), School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Claire Patterson
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - David Owen
- Starpharma Pty Ltd., 4-6 Southampton Cresent, Abbotsford, Victoria 3067, Australia
| | - Marianne Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Nadim Akhtar
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
93
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
94
|
Radwan Y, Arafa KK, Ghoniem MG, Al-Farraj ES, El-Sherbiny IM. An in-vitro quantitative investigation on the synergistic effect of capsaicin and 5-fluorouracil encapsulated into lipid nanocapsules to treat breast cancer. Drug Dev Ind Pharm 2023; 49:271-280. [PMID: 37067846 DOI: 10.1080/03639045.2023.2203252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Background: Breast cancer conventional therapeutics are effective; however, they encounter some limitations including multidrug resistance, the presence of pharmacological barriers and non-selectivity which hinder their optimal therapeutic efficacy. Aim: Overcoming such drawbacks necessitates the development of efficient drug vehicles including lipid-based nanoparticles. This study aimed to quantitatively investigate in-vitro the synergistic therapeutic effect of the novel combination of capsaicin and 5-fluorouracil (5-FU) encapsulated in lipid nanocapsules (LNCs). Method: To this end, thorough physicochemical and in-vitro assessments on breast cancer cell line (MCF-7) were done. The drugs-loaded LNCs were characterized using DLS, TEM imaging, stability study, and in-vitro release study. Results: The developed nanoformulations were monodisperse with average particle size (PS) of 31, 43.8 and 127.3 nm for empty LNCs, Cap-LNCs, and 5-FU-LNCs, respectively, and with a surface charge of -35.4, -21.7 and -31.4 mV, respectively, reflecting good physical stability. The TEM micrographs revealed the spherical morphology of the drugs-loaded LNCs with comparable PS to that obtained by DLS. Furthermore, the biological activity of the prepared LNCs were assessed through implementing comparative cytotoxicity studies as well as apoptosis, and cell cycle flow cytometric analyses. All biological assessments confirmed the superior antiproliferative effect of the combined drug-loaded LNCs over their free drugs counterparts. Conclusion: Intriguingly, the study findings highlighted the potential synergistic activity of the drugs (capsaicin and 5-FU) and the extensive enhancement of their biological activity through incorporation into LNCs. Promising results will pave the way to further assess the novel combined nanoformulation in preclinical and clinical studies on breast cancer patients.
Collapse
Affiliation(s)
- Yasmine Radwan
- Nanomedicine Research Labs, Center for Materials Sciences, Zewail City of Science and Technology, 6th of October City, 12578, Giza, Egypt
| | - Kholoud K Arafa
- Nanomedicine Research Labs, Center for Materials Sciences, Zewail City of Science and Technology, 6th of October City, 12578, Giza, Egypt
| | - Monira G Ghoniem
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Eida S Al-Farraj
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Sciences, Zewail City of Science and Technology, 6th of October City, 12578, Giza, Egypt
| |
Collapse
|
95
|
Zhang P, Zhu Y, Xiao C, Chen X. Activatable dual-functional molecular agents for imaging-guided cancer therapy. Adv Drug Deliv Rev 2023; 195:114725. [PMID: 36754284 DOI: 10.1016/j.addr.2023.114725] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 01/16/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
Theranostics has attracted great attention due to its ability to combine the real-time diagnosis of cancers with efficient treatment modalities. Activatable dual-functional molecular agents could be synthesized by covalently conjugating imaging agents, therapeutic agents, stimuli-responsive linkers and/or targeting molecules together. They could be selectively activated by overexpressed physiological stimuli or external triggers at the tumor sites to release imaging agents and cytotoxic drugs, thus offering many advantages for tumor imaging and therapy, such as a high signal-to-noise ratio, low systemic toxicity, and improved therapeutic effects. This review summarizes the recent advances of dual-functional molecular agents that respond to various physiological or external stimuli for cancer theranostics. The molecular designs, synthetic strategies, activatable mechanisms, and biomedical applications of these molecular agents are elaborated, followed by a brief discussion of the challenges and opportunities in this field.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China; State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200433, PR China
| | - Yaowei Zhu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| |
Collapse
|
96
|
Zein nanoparticles for drug delivery: Preparation methods and biological applications. Int J Pharm 2023; 635:122754. [PMID: 36812950 DOI: 10.1016/j.ijpharm.2023.122754] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Zein, a vegetable protein extracted from corn (Zea mays L.), forms a gastro-resistant and mucoadhesive polymer that is cheap and easy to obtain and facilitates the encapsulation of bioactives with hydrophilic, hydrophobic, and amphiphilic properties. The methods used for synthesizing these nanoparticles include antisolvent precipitation/nanoprecipitation, pH-driven, electrospraying, and solvent emulsification-evaporation methods. Each method has its advantages in the preparation of nanocarriers, nevertheless, all of them enable the production of zein nanoparticles that are stable and resistant to environmental factors, with different biological activities required in the cosmetic, food, and pharmaceutical industries. Therefore, zein nanoparticles are promising nanocarriers that can encapsulate various bioactives with anti-inflammatory, antioxidant, antimicrobial, anticancer, and antidiabetic properties. This article reviews the principal methods for obtaining zein nanoparticles containing bioactives, the advantages and characteristics of each method, as well as the main biological applications of nanotechnology-based formulations.
Collapse
|
97
|
Yusuf A, Almotairy ARZ, Henidi H, Alshehri OY, Aldughaim MS. Nanoparticles as Drug Delivery Systems: A Review of the Implication of Nanoparticles' Physicochemical Properties on Responses in Biological Systems. Polymers (Basel) 2023; 15:polym15071596. [PMID: 37050210 PMCID: PMC10096782 DOI: 10.3390/polym15071596] [Citation(s) in RCA: 216] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
In the last four decades, nanotechnology has gained momentum with no sign of slowing down. The application of inventions or products from nanotechnology has revolutionised all aspects of everyday life ranging from medical applications to its impact on the food industry. Nanoparticles have made it possible to significantly extend the shelf lives of food product, improve intracellular delivery of hydrophobic drugs and improve the efficacy of specific therapeutics such as anticancer agents. As a consequence, nanotechnology has not only impacted the global standard of living but has also impacted the global economy. In this review, the characteristics of nanoparticles that confers them with suitable and potentially toxic biological effects, as well as their applications in different biological fields and nanoparticle-based drugs and delivery systems in biomedicine including nano-based drugs currently approved by the U.S. Food and Drug Administration (FDA) are discussed. The possible consequence of continuous exposure to nanoparticles due to the increased use of nanotechnology and possible solution is also highlighted.
Collapse
Affiliation(s)
- Azeez Yusuf
- Irish Centre for Genetic Lung Disease, Department of Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, D02 YN77 Dublin, Ireland
| | | | - Hanan Henidi
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Ohoud Y Alshehri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11564, Saudi Arabia
| | - Mohammed S Aldughaim
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 11451, Saudi Arabia
| |
Collapse
|
98
|
Hasani M, Jafari S, Akbari Javar H, Abdollahi H, Rashidzadeh H. Cell-Penetrating Peptidic GRP78 Ligand-Conjugated Iron Oxide Magnetic Nanoparticles for Tumor-Targeted Doxorubicin Delivery and Imaging. ACS APPLIED BIO MATERIALS 2023; 6:1019-1031. [PMID: 36862384 DOI: 10.1021/acsabm.2c00897] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Although chemotherapy is regarded as an essential option in cancer treatment, it is still far from being perfect. Inadequate tumor drug concentration and systemic toxicity along with broad biodistribution have diminished the utility of chemotherapy. Tumor-targeting peptide-conjugated multifunctional nanoplatforms have emerged as an effective strategy for site-directed tumor tissues in cancer treatment and imaging. Herein, Pep42-targeted iron oxide magnetic nanoparticles (IONPs) functionalized with β-cyclodextrin (ßCD) containing doxorubicin (DOX) (Fe3O4-ßCD-Pep42-DOX) were successfully developed. The physical effects of the prepared NPs were characterized by employing various techniques. Transmission electron microscopy (TEM) images disclosed that the developed Fe3O4-ßCD-Pep42-DOX nanoplatforms had a spherical morphology and a core-shell structure with a size of nearly 17 nm. Fourier transform infrared (FT-IR) spectroscopy showed that β-cyclodextrin, DOX, and Pep42 molecules were successfully loaded on the IONPs. In vitro cytotoxicity analysis revealed that the fabricated multifunctional Fe3O4-ßCD-Pep42 nanoplatforms possessed excellent biosafety toward BT-474, MDA-MB468 (cancerous cells), and MCF10A normal cells, while Fe3O4-ßCD-Pep42-DOX exhibited great cancer cell killing ability. The high cellular uptake along with intracellular trafficking of Fe3O4-ßCD-Pep42-DOX highlights the usefulness of the Pep42-targeting peptide. In vivo results strongly supported the in vitro results, i.e., significant tumor size reduction was observed by single-dose injection of Fe3O4-ßCD-Pep42-DOX into tumor-bearing mice. Interestingly, in vivo MR imaging (MRI) of Fe3O4-ßCD-Pep42-DOX revealed T2 contrast improvement in the tumor cells and therapeutic ability in cancer theranostics. Taken together, these findings provided strong evidence for the potential capability of Fe3O4-ßCD-Pep42-DOX as a multifunctional nanoplatform in cancer therapy and imaging and opens up a new avenue of research in this area.
Collapse
Affiliation(s)
- Mahdiyeh Hasani
- Pharmaceutical Nanotechnology Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Zanjan University of Medical Sciences, Zanjan PG36+6RX, Iran
| | - Samira Jafari
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah 83VX+PCM, Iran
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran P94V+8MF, Iran
| | - Hossein Abdollahi
- Department of Polymer Engineering, Faculty of Engineering, Urmia University, Urmia 5756151818, Iran
| | - Hamid Rashidzadeh
- Pharmaceutical Nanotechnology Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Zanjan University of Medical Sciences, Zanjan PG36+6RX, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan PG36+6RX, Iran
| |
Collapse
|
99
|
Haq Khan ZU, Khan TM, Khan A, Shah NS, Muhammad N, Tahir K, Iqbal J, Rahim A, Khasim S, Ahmad I, Shabbir K, Gul NS, Wu J. Brief review: Applications of nanocomposite in electrochemical sensor and drugs delivery. Front Chem 2023; 11:1152217. [PMID: 37007050 PMCID: PMC10060975 DOI: 10.3389/fchem.2023.1152217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
The recent advancement of nanoparticles (NPs) holds significant potential for treating various ailments. NPs are employed as drug carriers for diseases like cancer because of their small size and increased stability. In addition, they have several desirable properties that make them ideal for treating bone cancer, including high stability, specificity, higher sensitivity, and efficacy. Furthermore, they might be taken into account to permit the precise drug release from the matrix. Drug delivery systems for cancer treatment have progressed to include nanocomposites, metallic NPs, dendrimers, and liposomes. Materials’ mechanical strength, hardness, electrical and thermal conductivity, and electrochemical sensors are significantly improved using nanoparticles (NPs). New sensing devices, drug delivery systems, electrochemical sensors, and biosensors can all benefit considerably from the NPs’ exceptional physical and chemical capabilities. Nanotechnology is discussed in this article from a variety of angles, including its recent applications in the medical sciences for the effective treatment of bone cancers and its potential as a promising option for treating other complex health anomalies via the use of anti-tumour therapy, radiotherapy, the delivery of proteins, antibiotics, and vaccines, and other methods. This also brings to light the role that model simulations can play in diagnosing and treating bone cancer, an area where Nanomedicine has recently been formulated. There has been a recent uptick in using nanotechnology to treat conditions affecting the skeleton. Consequently, it will pave the door for more effective utilization of cutting-edge technology, including electrochemical sensors and biosensors, and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Zia Ul Haq Khan
- Department of Environmental Sciences, COMSATS University Islamabad, Vehari, Pakistan
- *Correspondence: Zia Ul Haq Khan, ; Noor Shad Gul,
| | - Taj Malook Khan
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Department of Pharmacology, Laboratory of Cardiovascular Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Amjad Khan
- Department of Zoology, University of Lakki Marwat, Lakki Marwat, Pakistan
| | - Noor Samad Shah
- Department of Environmental Sciences, COMSATS University Islamabad, Vehari, Pakistan
| | - Nawshad Muhammad
- Department of Dental Materials, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Kamran Tahir
- Institute of Chemical Sciences, Gomal University, Dera Ismail Khan, Pakistan
| | - Jibran Iqbal
- College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Abdur Rahim
- Department of Chemistry, COMSATS University Islamabad, Islamabad, Pakistan
| | - Syed Khasim
- Nanotechnology Research Unit, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
- Department of Physics, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Iftikhar Ahmad
- Department of Environmental Sciences, COMSATS University Islamabad, Vehari, Pakistan
| | - Khadija Shabbir
- Department of Environmental Sciences, COMSATS University Islamabad, Vehari, Pakistan
| | - Noor Shad Gul
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Department of Pharmacology, Laboratory of Cardiovascular Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
- *Correspondence: Zia Ul Haq Khan, ; Noor Shad Gul,
| | - Jianbo Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Department of Pharmacology, Laboratory of Cardiovascular Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
100
|
Witkowska M, Golusińska-Kardach E, Golusiński W, Florek E. Polydopamine-Based Material and Their Potential in Head and Neck Cancer Therapy-Current State of Knowledge. Int J Mol Sci 2023; 24:4890. [PMID: 36902321 PMCID: PMC10003234 DOI: 10.3390/ijms24054890] [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: 02/14/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Head and neck cancers (HNC) are among the most common cancers in the world. In terms of frequency of occurrence in the world, HNC ranks sixth. However, the problem of modern oncology is the low specificity of the therapies used, which is why most of the currently used chemotherapeutic agents have a systemic effect. The use of nanomaterials could overcome the limitations of traditional therapies. Researchers are increasingly using polydopamine (PDA) in nanotherapeutic systems for HNC due to its unique properties. PDA has found applications in chemotherapy, photothermal therapy, targeted therapy, and combination therapies that facilitate better carrier control for the effective reduction of cancer cells than individual therapies. The purpose of this review was to present the current knowledge on the potential use of polydopamine in head and neck cancer research.
Collapse
Affiliation(s)
- Marta Witkowska
- Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznan, Poland
- Centre for Advanced Technologies, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Ewelina Golusińska-Kardach
- Department and Clinic of Dental Surgery, Periodontal Diseases and Oral Mucosa, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Wojciech Golusiński
- Department and Clinic of Head and Neck Surgery and Laryngological Oncology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
| | - Ewa Florek
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| |
Collapse
|