1
|
Kaplan Ö, Gök MK, Pekmez M, Erden Tayhan S, Özgümüş S, Gökçe İ, Arda N. Development of recombinant protein-based nanoparticle systems for inducing tumor cell apoptosis: In vitro evaluation of their cytotoxic and apoptotic effects on cancer cells. J Drug Deliv Sci Technol 2024; 95:105565. [DOI: 10.1016/j.jddst.2024.105565] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Dzhumashev D, Anton-Joseph S, Morel VJ, Timpanaro A, Bordon G, Piccand C, Aleandri S, Luciani P, Rössler J, Bernasconi M. Rapid liposomal formulation for nucleolin targeting to rhabdomyosarcoma cells. Eur J Pharm Biopharm 2024; 194:49-61. [PMID: 38029941 DOI: 10.1016/j.ejpb.2023.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. More effective and less toxic therapies are urgently needed for high-risk patients. Peptide-guided targeted drug delivery can increase the therapeutic index of encapsulated drugs and improve patients' well-being. To apply this strategy to RMS, we identified the peptide F3 in a screening for peptides binding to RMS cells surface. F3 binds to nucleolin, which is present on the surface of RMS cells and is abundantly expressed at the mRNA level in RMS patients' biopsies compared to healthy tissues. We developed a rapid microfluidic formulation of F3-decorated PEGylated liposomes and remote loading of the chemotherapeutic drug vincristine. Size, surface charge, drug loading and retention of targeted and control liposomes were studied. Enhanced cellular binding and uptake were observed in three different nucleolin-positive RMS cell lines. Importantly, F3-functionalized liposomes loaded with vincristine were up to 11 times more cytotoxic than non-targeted liposomes for RMS cell lines. These results demonstrate that F3-functionalized liposomes are promising for targeted drug delivery to RMS and warrant further in vivo investigations.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Stenija Anton-Joseph
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Victoria J Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Caroline Piccand
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
3
|
Raza F, Zheng M, Zhong H, Su J, He B, Yuan WE, Qiu M. Engineered tumor microvesicles modified by SP94 peptide for arsenic trioxide targeting drug delivery in liver cancer therapy. BIOMATERIALS ADVANCES 2023; 155:213683. [PMID: 37925825 DOI: 10.1016/j.bioadv.2023.213683] [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: 07/25/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Liver cancer is among the leading cause of cancer related death worldwide. There is growing interest in using traditional Chinese medicines such as arsenic trioxide (ATO) to treat liver cancer. ATO have attracted attention due to its wide range of anti-cancer activities. However, the current ATO formulations are associated with drawbacks such as short half-life, lack of targeting ability towards solid tumors and apparent toxic side effects. Tumor microvesicles (TMVs) has shown encouraging results for the delivery of drugs to solid tumor. In this work, we designed ATO loaded TMVs further modified by SP94 peptide as liver cancer specific ligand (ATO@SP94-TMVs). This drug delivery system utilized SP94 peptide that selectively targets liver cancer cells while TMVs increase the accumulation of ATO at tumor site and activate immune response owing to the associated antigens. ATO@SP94-TMVs exhibited high encapsulation efficiency and tumor microenvironment triggered enhanced release of ATO in vitro. Cytotoxicity and uptake studies revealed remarkable inhibition and specific targeting of H22 cells. In addition, excellent immune response was detected in vitro, enhancing anti-tumor efficacy. Furthermore, a tumor inhibition rate of about 53.23 % was observed in H22 bearing tumor model. Overall, these results confirm that ATO@SP94-TMVs can be a promising nano drug delivery system for the future liver cancer therapy and improve its clinical applications.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mengyuan Zheng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongyu Zhong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Beixuan He
- Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
4
|
Ma S, Kim JH, Chen W, Li L, Lee J, Xue J, Liu Y, Chen G, Tang B, Tao W, Kim JS. Cancer Cell-Specific Fluorescent Prodrug Delivery Platforms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207768. [PMID: 37026629 PMCID: PMC10238224 DOI: 10.1002/advs.202207768] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Indexed: 06/04/2023]
Abstract
Targeting cancer cells with high specificity is one of the most essential yet challenging goals of tumor therapy. Because different surface receptors, transporters, and integrins are overexpressed specifically on tumor cells, using these tumor cell-specific properties to improve drug targeting efficacy holds particular promise. Targeted fluorescent prodrugs not only improve intracellular accumulation and bioavailability but also report their own localization and activation through real-time changes in fluorescence. In this review, efforts are highlighted to develop innovative targeted fluorescent prodrugs that efficiently accumulate in tumor cells in different organs, including lung cancer, liver cancer, cervical cancer, breast cancer, glioma, and colorectal cancer. The latest progress and advances in chemical design and synthetic considerations in fluorescence prodrug conjugates and how their therapeutic efficacy and fluorescence can be activated by tumor-specific stimuli are reviewed. Additionally, novel perspectives are provided on strategies behind engineered nanoparticle platforms self-assembled from targeted fluorescence prodrugs, and how fluorescence readouts can be used to monitor the position and action of the nanoparticle-mediated delivery of therapeutic agents in preclinical models. Finally, future opportunities for fluorescent prodrug-based strategies and solutions to the challenges of accelerating clinical translation for the treatment of organ-specific tumors are proposed.
Collapse
Affiliation(s)
- Siyue Ma
- The Youth Innovation Team of Shaanxi UniversitiesShaanxi Key Laboratory of Chemical Additives for IndustryCollege of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'an710021China
- Key Laboratory of Emergency and Trauma, Ministry of EducationCollege of Emergency and TraumaHainan Medical UniversityHaikou571199China
| | - Ji Hyeon Kim
- Department of ChemistryKorea UniversitySeoul02841South Korea
| | - Wei Chen
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Lu Li
- The Youth Innovation Team of Shaanxi UniversitiesShaanxi Key Laboratory of Chemical Additives for IndustryCollege of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'an710021China
| | - Jieun Lee
- Department of ChemistryKorea UniversitySeoul02841South Korea
| | - Junlian Xue
- The Youth Innovation Team of Shaanxi UniversitiesShaanxi Key Laboratory of Chemical Additives for IndustryCollege of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'an710021China
| | - Yuxia Liu
- The Youth Innovation Team of Shaanxi UniversitiesShaanxi Key Laboratory of Chemical Additives for IndustryCollege of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'an710021China
| | - Guang Chen
- The Youth Innovation Team of Shaanxi UniversitiesShaanxi Key Laboratory of Chemical Additives for IndustryCollege of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'an710021China
- College of ChemistryChemical Engineering and Materials ScienceKey Laboratory of Molecular and Nano ProbesMinistry of EducationCollaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of ShandongInstitutes of Biomedical SciencesShandong Normal UniversityJinan250014China
| | - Bo Tang
- College of ChemistryChemical Engineering and Materials ScienceKey Laboratory of Molecular and Nano ProbesMinistry of EducationCollaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of ShandongInstitutes of Biomedical SciencesShandong Normal UniversityJinan250014China
| | - Wei Tao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Jong Seung Kim
- Department of ChemistryKorea UniversitySeoul02841South Korea
| |
Collapse
|
5
|
Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J Nanobiotechnology 2022; 20:109. [PMID: 35248080 PMCID: PMC8898455 DOI: 10.1186/s12951-022-01309-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Liver cancer is considered one of the deadliest diseases with one of the highest disease burdens worldwide. Among the different types of liver cancer, hepatocellular carcinoma is considered to be the most common type. Multiple conventional approaches are being used in treating hepatocellular carcinoma. Focusing on drug treatment, regular agents in conventional forms fail to achieve the intended clinical outcomes. In order to improve the treatment outcomes, utilizing nanoparticles-specifically lipid based nanoparticles-are considered to be one of the most promising approaches being set in motion. Multiple forms of lipid based nanoparticles exist including liposomes, solid lipid nanoparticles, nanostructured lipid carriers, microemulsion, nanoemulsion, phytosomes, lipid coated nanoparticles, and nanoassemblies. Multiple approaches are used to enhance the tumor uptake as well tumor specificity such as intratumoral injection, passive targeting, active targeting, and stimuli responsive nanoparticles. In this review, the effect of utilizing lipidic nanoparticles is being discussed as well as the different tumor uptake enhancement techniques used.
Collapse
Affiliation(s)
- Khaled Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
| | - Shady Swidan
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
| | - Mohamed El-Nabarawi
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Mahmoud Teaima
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
6
|
Ladju RB, Ulhaq ZS, Soraya GV. Nanotheranostics: A powerful next-generation solution to tackle hepatocellular carcinoma. World J Gastroenterol 2022; 28:176-187. [PMID: 35110943 PMCID: PMC8776531 DOI: 10.3748/wjg.v28.i2.176] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/15/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is an epidemic burden and remains highly prevalent worldwide. The significant mortality rates of HCC are largely due to the tendency of late diagnosis and the multifaceted, complex nature of treatment. Meanwhile, current therapeutic modalities such as liver resection and transplantation are only effective for resolving early-stage HCC. Hence, alternative approaches are required to improve detection and enhance the efficacy of current treatment options. Nanotheranostic platforms, which utilize biocompatible nanoparticles to perform both diagnostics and targeted delivery, has been considered a potential approach for cancer management in the past few decades. Advancement of nanomaterials and biomedical engineering techniques has led to rapid expansion of the nanotheranostics field, allowing for more sensitive and specific diagnosis, real-time monitoring of drug delivery, and enhanced treatment efficacies across various malignancies. The focus of this review is on the applications of nanotheranostics for HCC. The review first explores the current epidemiology and the commonly encountered obstacles in HCC diagnosis and treatment. It then presents the current technological and functional advancements in nanotheranostic technology for cancer in general, and then specifically explores the use of nanotheranostic modalities as a promising option to address the key challenges present in HCC management.
Collapse
Affiliation(s)
- Rusdina Bte Ladju
- Department of Anatomic Pathology, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| | - Zulvikar Syambani Ulhaq
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim Islamic State University, Malang 65151, Indonesia
- National Research and Innovation Agency, Central Jakarta 10340, Indonesia
| | - Gita Vita Soraya
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
- Department of Neurology, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia
| |
Collapse
|
7
|
Li T, Chen X, Wan J, Hu X, Chen W, Wang H. Akt inhibition improves the efficacy of cabazitaxel nanomedicine in preclinical taxane-resistant cancer models. Int J Pharm 2021; 607:121017. [PMID: 34416334 DOI: 10.1016/j.ijpharm.2021.121017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/18/2021] [Accepted: 08/15/2021] [Indexed: 12/20/2022]
Abstract
Drug resistance remains a major challenge in achieving cures in cancer patients. Cabazitaxel has shown the ability to overcome drug resistance induced by paclitaxel and docetaxel; however, substantially high toxicity has been observed in patients receiving this agent, which compromises its efficacy. We have previously demonstrated that a polymeric platform (termed cabazitaxel-NPs) encapsulating the oligolactide-cabazitaxel conjugate exhibits desired antitumor efficacy and improved in vivo tolerability. However, we found that upon cabazitaxel treatment, cancer cells adapted to activate Akt signaling, which potentially discounts the drug efficacy. We therefore hypothesized that combing cabazitaxel nanotherapeutics with a pan-Akt inhibitor MK-2206 would synergistically sensitize the resistant cancer. In this study, we confirmed that nanoparticle formulation reduced the systemic toxicity, with higher tolerance than solution-based free cabazitaxel agent in animals. Interestingly, the activation of Akt signaling in the resistant cancer was reversed by the addition of MK-2206. In particular, the collaboration of these two ingredients was demonstrated to maximize the efficacy in vitro and in a xenograft model bearing paclitaxel-resistant tumors. Mechanistically, Akt inhibition increased the microtubule-stabilizing effect of cabazitaxel nanomedicine. Collectively, this report introduced a binary platform composed of cytotoxic nanotherapeutics and inhibitors with certain targets to combat multidrug resistance, and such a combined regimen has the potential for the clinical treatment of patients with resistant cancer.
Collapse
Affiliation(s)
- Tongyu Li
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, PR China
| | - Xiaona Chen
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, PR China
| | - Jianqin Wan
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, PR China
| | - Xiaoxiao Hu
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, PR China
| | - Wanzhi Chen
- Department of Chemistry, Zhejiang University, Hangzhou 310028, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, PR China.
| |
Collapse
|
8
|
Li Z, Wang F, Li Y, Wang X, Lu Q, Wang D, Qi C, Li C, Li Z, Lian B, Tian G, Gao Z, Zhang B, Wu J. Combined anti-hepatocellular carcinoma therapy inhibit drug-resistance and metastasis via targeting "substance P-hepatic stellate cells-hepatocellular carcinoma" axis. Biomaterials 2021; 276:121003. [PMID: 34273686 DOI: 10.1016/j.biomaterials.2021.121003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Peripheral nerves have emerged as the important components in tumor microenvironment (TME), which could activate hepatic stellate cells (HSCs) by secreting substance P (SP), leading to hepatocellular carcinoma (HCC) invasion and metastasis. Herein, we proposed a novel anti-HCC concept of blocking "SP-HSCs-HCC" axis for omnidirectional inhibition of HCC development. To pursue this aim, the novel CAP/GA-sHA-DOX NPs were developed for targeted co-delivery of capsaicin (CAP) and doxorubicin (DOX) using glycyrrhetinic acid (GA) modified sulfated-HA (sHA) as nanocarriers. Among that, CAP could inhibit the activation of HSCs as an inhibitor of SP. Notably, to real mimic "SP-HSCs-HCC" axis for in vitro and in vivo evaluation, both "SP + LX-2+BEL-7402" co-cultured cell model and "SP + m-HSC + H22" co-implantation mice model were attempted for the first time. Furthermore, in vivo anti-HCC effects were performed in three different tumor-bearing models: subcutaneous implantation of H22 or "SP + m-HSC + H22", intravenous injection of H22 for lung metastasis, and orthotopic implantation of H22 for primary HCC. Our results showed that CAP/GA-sHA-DOX NPs could be efficiently taken up by tumor cells and activated HSCs (aHSCs) simultaneously, and effectively inhibit tumor drug-resistance and migration by blocking SP-induced HSCs activation. In addition, CAP/GA-sHA-DOX NPs exhibited low ECM deposition, less tumor angiogenesis, and superior in vivo anti-HCC effects. The anti-HCC mechanisms revealed that CAP/GA-sHA-DOX NPs could down-regulate the expression level of Vimentin and P-gp, reverse epithelial-mesenchymal transition (EMT) of tumor cells. In brief, the nano-sized combination therapy based on GA-sHA-DOX polymers could effectively inhibit drug-resistance and metastasis of HCC by blocking "SP-HSCs-HCC" axis, which provides a promising approach for cancer therapy.
Collapse
Affiliation(s)
- Zhipeng Li
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Fangqing Wang
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Yanying Li
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Xiaoxue Wang
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Qiao Lu
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Di Wang
- School of Nursing, Weifang Medical University, PR China
| | - Cuiping Qi
- School of Nursing, Weifang Medical University, PR China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China
| | - Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Guixiang Tian
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Zhiqin Gao
- School of Bioscience and Technology, Weifang Medical University, PR China.
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China.
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, PR China.
| |
Collapse
|
9
|
Niza E, Ocaña A, Castro-Osma JA, Bravo I, Alonso-Moreno C. Polyester Polymeric Nanoparticles as Platforms in the Development of Novel Nanomedicines for Cancer Treatment. Cancers (Basel) 2021; 13:3387. [PMID: 34298604 PMCID: PMC8304499 DOI: 10.3390/cancers13143387] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
Many therapeutic agents have failed in their clinical development, due to the toxic effects associated with non-transformed tissues. In this context, nanotechnology has been exploited to overcome such limitations, and also improve navigation across biological barriers. Amongst the many materials used in nanomedicine, with promising properties as therapeutic carriers, the following one stands out: biodegradable and biocompatible polymers. Polymeric nanoparticles are ideal candidates for drug delivery, given the versatility of raw materials and their feasibility in large-scale production. Furthermore, polymeric nanoparticles show great potential for easy surface modifications to optimize pharmacokinetics, including the half-life in circulation and targeted tissue delivery. Herein, we provide an overview of the current applications of polymeric nanoparticles as platforms in the development of novel nanomedicines for cancer treatment. In particular, we will focus on the raw materials that are widely used for polymeric nanoparticle generation, current methods for formulation, mechanism of action, and clinical investigations.
Collapse
Affiliation(s)
- Enrique Niza
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Alberto Ocaña
- Experimental Therapeutics Unit, Hospital Clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain;
| | - José Antonio Castro-Osma
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain; (E.N.); (J.A.C.-O.)
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
10
|
Abdel-Rahman SA, Wafa EI, Ebeid K, Geary SM, Naguib YW, El-Damasy AK, Salem AK. Thiophene derivative-loaded nanoparticles mediate anticancer activity through the inhibition of kinases and microtubule assembly. ADVANCED THERAPEUTICS 2021; 4. [PMID: 34423112 DOI: 10.1002/adtp.202100058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Different tetrahydrobenzo[b]thiophene derivatives were explored as new tubulin polymerization destabilizers to arrest tumor cell mitosis. A series of compounds incorporating the tetrahydrobenzo[b]thiophene scaffold were synthesized, and their biological activities were investigated. The cytotoxicity of each of the synthesized compounds was assessed against a range of cell lines. Specifically, the benzyl urea tetrahydrobenzo[b]thiophene derivative, 1-benzyl-3-(3-cyano-4,5,6,7-tetrahydrobenzo[b]thiophen-2-yl)urea (BU17), was identified as the most potent compound with broad-spectrum antitumor activity against several cancer cell lines. The potential mechanism(s) of action were investigated where dose-dependent G2/M accumulation and A549 cell cycle arrest were detected. Additionally, A549 cells treated with BU17 expressed enhanced levels of caspase 3 and 9, indicating the induction of apoptosis. Furthermore, it was found that BU17 inhibits WEE1 kinase and targets tubulin by blocking its polymerization. BU17 was also formulated into PLGA nanoparticles, and it was demonstrated that BU17-loaded nanoparticles could significantly enhance antitumor activity compared to the soluble counterpart.
Collapse
Affiliation(s)
- Somaya A Abdel-Rahman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.,Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.,Department of Pharmaceutics, Faculty of Pharmacy, Deraya University, New Minia City, Minia, 61519 Egypt
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.,Department of Pharmaceutics, Faculty of Pharmacy, Deraya University, New Minia City, Minia, 61519 Egypt
| | - Ashraf K El-Damasy
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
11
|
Ding N, Xu S, Zheng S, Ye Q, Xu L, Ling S, Xie S, Chen W, Zhang Z, Xue M, Lin Z, Xu X, Wang L. "Sweet tooth"-oriented SN38 prodrug delivery nanoplatform for targeted gastric cancer therapy. J Mater Chem B 2021; 9:2816-2830. [PMID: 33690741 DOI: 10.1039/d0tb02787a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Most cancer cells employ overexpression of glucose transports (GLUTs) to satisfy glucose demand ("Sweet Tooth") for increased aerobic glycolysis rates. GLUT1, one of the most widely expressed GLUTs in numerous cancers, was identified as a prognosis-related biomarker of gastric cancer via tissue array analysis. Herein, a "Sweet Tooth"-oriented SN38 prodrug delivery nanoplatform (Glu-SNP) was developed for targeted gastric cancer therapy. For this purpose, a SN38-derived prodrug (PLA-SN38) was synthesized by tethering 7-ethyl-10-hydroxycamptothecin (SN38) to biocompatible polylactic acid (PLA) with the appropriate degree of polymerization (n = 44). The PLA-SN38 conjugate was further assembled with glycosylated amphiphilic lipid to obtain glucosamine-decorated nanoparticles (Glu-SNP). Glu-SNP exhibited potent antitumor efficiency both in vitro and in vivo through enhanced cancer cell-specific targeting associated with the overexpression of GLUT1, which provides a promising approach for gastric cancer therapy.
Collapse
Affiliation(s)
- Ning Ding
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310020, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fang T, Ye Z, Chen X, Wang Y, Wan J, Wang H. Repurposing of camptothecin: An esterase-activatable prodrug delivered by a self-emulsifying formulation that improves efficacy in colorectal cancer. Int J Pharm 2021; 599:120399. [PMID: 33647408 DOI: 10.1016/j.ijpharm.2021.120399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/27/2021] [Accepted: 02/14/2021] [Indexed: 01/05/2023]
Abstract
The global burden of colorectal cancer (CRC), the third most commonly diagnosed malignancy, continues to rise. Therefore, more effective and less toxic therapies are needed for CRC. CPT-11 (also called irinotecan), the standard-of-care treatment for CRC, has only had limited effects on survival outcomes. In vivo, CPT-11 must be converted to an active metabolite, SN38, to exert antitumor activity in the presence of carboxylesterases, but the conversion rate is extremely low (usually less than 8%). To fully harness the active SN38 compound, we showed here that esterification of SN38 using α-linolenic acid (LNA) generated a prodrug (termed LSN38), which can be formulated in pharmaceutically acceptable surfactants, such as polysorbate 80. Upon blending with an aqueous ethanolic solution, the mixture of LSN38/polysorbate 80 formed self-emulsifying nanomicelles (termed LSN38 NMs), enabling systemic injection. Unlike the insufficient release of active SN38 from CPT-11, drug activation from the LSN38 prodrug was quantitative and relied on esterase, which is abundant in cancerous cells. Pharmacokinetics studies revealed that polysorbate 80-based nanomicelles stably constrained the prodrug in the reservoir and prolonged blood circulation compared to CPT-11. Furthermore, LSN38 NMs showed superior therapeutic efficacy against a colorectal xenograft-bearing mouse model that failed to be treated with clinically approved CPT-11. Overall, these studies highlight the feasibility of converting a chemotherapeutic agent that is not miscible or compatible with pharmaceutical surfactants into an injectable self-emulsifying formulation. This approach could be applied to rescue other drugs or drug candidates that are abandoned in the preclinical stages due to pharmaceutical challenges.
Collapse
Affiliation(s)
- Tao Fang
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, PR China
| | - Zhijian Ye
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, PR China
| | - Xiaona Chen
- The First Affiliated Hospital, Zhejiang University School of Medicine, NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, PR China
| | - Yuchen Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, PR China
| | - Jianqin Wan
- The First Affiliated Hospital, Zhejiang University School of Medicine, NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine, NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, PR China.
| |
Collapse
|
13
|
Enhanced In Vitro Magnetic Cell Targeting of Doxorubicin-Loaded Magnetic Liposomes for Localized Cancer Therapy. NANOMATERIALS 2020; 10:nano10112104. [PMID: 33114052 PMCID: PMC7690690 DOI: 10.3390/nano10112104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
The lack of efficient targeting strategies poses significant limitations on the effectiveness of chemotherapeutic treatments. This issue also affects drug-loaded nanocarriers, reducing nanoparticles cancer cell uptake. We report on the fabrication and in vitro characterization of doxorubicin-loaded magnetic liposomes for localized treatment of liver malignancies. Colloidal stability, superparamagnetic behavior and efficient drug loading of our formulation were demonstrated. The application of an external magnetic field guaranteed enhanced nanocarriers cell uptake under cell medium flow in correspondence of a specific area, as we reported through in vitro investigation. A numerical model was used to validate experimental data of magnetic targeting, proving the possibility of accurately describing the targeting strategy and predict liposomes accumulation under different environmental conditions. Finally, in vitro studies on HepG2 cancer cells confirmed the cytotoxicity of drug-loaded magnetic liposomes, with cell viability reduction of about 50% and 80% after 24 h and 72 h of incubation, respectively. Conversely, plain nanocarriers showed no anti-proliferative effects, confirming the formulation safety. Overall, these results demonstrated significant targeting efficiency and anticancer activity of our nanocarriers and superparamagnetic nanoparticles entrapment could envision the theranostic potential of the formulation. The proposed magnetic targeting study could represent a valid tool for pre-clinical investigation regarding the effectiveness of magnetic drug targeting.
Collapse
|
14
|
Chen C, Zhou L, Xie B, Wang Y, Ren L, Chen X, Cen B, Lv H, Wang H. Novel fast-acting pyrazole/pyridine-functionalized N-heterocyclic carbene silver complexes assembled with nanoparticles show enhanced safety and efficacy as anticancer therapeutics. Dalton Trans 2020; 49:2505-2516. [PMID: 32022055 DOI: 10.1039/c9dt04751d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In this study, we designed and synthesized four novel multi-nuclear silver complexes (1-4) coordinated with pyrazole- or pyridine-functionalized N-heterocyclic carbene (NHC) ligands. The crystal structures of the silver-NHC complexes were confirmed by X-ray diffraction analysis. In vitro assays showed that the silver-NHC complexes effectively killed a broad range of cancer cells after short-term drug exposure, serving as fast-acting cytotoxic agents. Of note, in cisplatin-resistant A549 cancer cells, the silver complexes were not cross-resistant with the clinically used cisplatin agent. Detailed mechanistic studies revealed that complex 2 triggered caspase-independent cell necrosis associated with intracellular reactive oxygen species (ROS) production and mitochondrial membrane potential (MMP) depletion. By exploiting a facile nano-assembly process, silver-NHC complexes 1, 2 and 4 were successfully integrated into the hydrophobic cores of amphiphilic matrices (DSPE-PEG2K), enabling systemic injection. The silver complex-loaded nanotherapeutics (1-NPs, 2-NPs, and 4-NPs) showed high safety margins with reduced systemic drug toxicities relative to cisplatin in animals. Furthermore, in a xenograft model of human colorectal cancer, the administration of the nanotherapeutics resulted in a marked inhibition of tumor progression.
Collapse
Affiliation(s)
- Chao Chen
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China. and College of Life Sciences, Huzhou University, Huzhou, 313000, PR China
| | - Liqian Zhou
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, PR China
| | - Yuchen Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| | - Lulu Ren
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, PR China
| | - Xiaona Chen
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| | - Beini Cen
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| | - He Lv
- College of Life Sciences, Huzhou University, Huzhou, 313000, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| |
Collapse
|
15
|
Yusheng S, Chenjun M, Yingying H, Tiantian W, Liefeng Z. Multifunctional nanoparticles of paclitaxel and cyclodextrin-polypeptide conjugates with in vitro anticancer activity. Pharm Dev Technol 2020; 25:1071-1080. [PMID: 32589088 DOI: 10.1080/10837450.2020.1787441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this study, the cyclodextrin polypeptide (R8-CMβCD) was successfully synthesized by the conjugation of a cell-penetrating peptide (R8) with carboxymethyl-β-cyclodextrin (CMβCD) via the carbon diamine reaction. Then, paclitaxel-loaded nanoparticles (PTX@R8-CMβCD NPs) was prepared. Results of transmission electron microscopy (TEM) showed that PTX@R8-CMβCD NPs were spherical with smooth surfaces and an average diameter about 144 nm. The amount of PTX released from NPs was less than 20% at pH7.4, but it increased significantly to 80% in the weakly acidic cytoplasm of tumors (pH5.0). Furthermore, PTX@R8-CMβCD NPs promoted the cellular uptake of PTX. Further studies on the mechanism showed that cellular uptake of PTX@R8-CMβCD NPs could rely on multiple pathways. In addition, the NPs had the ability to inhibit P-gp efflux pumps. Cytotoxicity tests showed that the NPs had no side effects. Taken together, PTX@R8-CMβCD NPs is an effective anticancer drug delivery system, and the material (R8-CMβCD) may be a promising anti-cancer drug carrier.
Collapse
Affiliation(s)
- Sun Yusheng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Ma Chenjun
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Hua Yingying
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Wei Tiantian
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Zhang Liefeng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
16
|
Bai YH, Yun XJ, Xue Y, Zhou T, Sun X, Gao YJ. A novel oncolytic adenovirus inhibits hepatocellular carcinoma growth. J Zhejiang Univ Sci B 2020; 20:1003-1013. [PMID: 31749347 DOI: 10.1631/jzus.b1900089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the inhibitory role of a novel oncolytic adenovirus (OA), GP73-SphK1sR-Ad5, on the growth of hepatocellular carcinoma (HCC). METHODS GP73-SphK1sR-Ad5 was constructed by integrating Golgi protein 73 (GP73) promoter and sphingosine kinase 1 (SphK1)-short hairpin RNA (shRNA) into adenovirus serotype 5 (Ad5), and transfecting into HCC Huh7 cells and normal human liver HL-7702 cells. The expression of SphK1 and adenovirus early region 1 (E1A) was detected by quantitative real-time PCR (qRT-PCR) and western blot, respectively. Cell viability was detected by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, and apoptotic rate was determined by flow cytometry. An Huh7 xenograft model was established in mice injected intratumorally with GP73-SphK1sR-Ad5. Twenty days after injection, the tumor volume and weight, and the survival time of the mice were recorded. The histopathological changes in tumor tissues were observed by hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM). RESULTS Transfection of GP73-SphK1sR-Ad5 significantly upregulated E1A and downregulated SphK1 in Huh7 cells, but not in HL7702 cells. GP73-SphK1sR-Ad5 transfection significantly decreased the viability and increased the apoptotic rate of Huh7 cells, but had no effect on HL7702 cells. Intratumoral injection of GP73-SphK1sR-Ad5 into the Huh7 xenograft mouse model significantly decreased tumor volume and weight, and prolonged survival time. It also significantly decreased the tumor infiltration area and blood vessel density, and increased the percentages of cells with nucleus deformation and cells with condensed chromatin in tumor tissues. CONCLUSIONS GP73-SphK1sR-Ad5 serves as a novel OA and can inhibit HCC progression with high specificity and efficacy.
Collapse
Affiliation(s)
- Yu-Huan Bai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China.,Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Xiao-Jing Yun
- Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Yan Xue
- Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Ting Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xin Sun
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yan-Jing Gao
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
17
|
Jefremow A, Neurath MF. Nanoparticles in Gastrooncology. Visc Med 2020; 36:88-94. [PMID: 32355665 PMCID: PMC7184848 DOI: 10.1159/000506908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastrointestinal malignancies have the greatest incidence and cancer-associated death rates worldwide. Routine therapeutic modalities include surgery, chemotherapy and radiation but they often fail to reach the goal of cancer-free survival. SUMMARY In the light of this urgent medical need for the treatment of GI tumors, nanotech-nology-based approaches, i.e. nanomedicine, promise new therapeutic options. Using nanoparticles instead of classically designed drugs, targeting anticancer agents directly to the tumor site may revolutionize both diagnostic and therapeutic tools thereby facilitating the identification and elimination of malignant cells. Importantly, diagnostic insight and therapeutic effects can be achieved simultaneously through the same nanoparticle. Additionally, a nanoparticle may be loaded with more than one agent, thereby further increasing the value and power of the nanotechnology approach in oncologic therapeutic concepts. Although most insight into mechanisms of nanomedicine has been gained from in vitro and preclinical in vivo models, few clinical trials have been conducted, and nanomedicine-based concepts are already part of standard treatment algorithms. However, despite substantial progress it remains a challenge to design nanoparticles that feature all desirable characteristics at the same time. KEY MESSAGES This review seeks to provide substantial insight into the current status of nanomedicine-based approaches employed for diagnostic and/or therapeutic purposes in the field of gastrointestinal cancers by highlighting achievements and pointing out unresolved issues that need to be further addressed by future research attempts.
Collapse
Affiliation(s)
| | - Markus F. Neurath
- Department of Internal Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
18
|
Tao Y, Wang J, Xu X. Emerging and Innovative Theranostic Approaches for Mesoporous Silica Nanoparticles in Hepatocellular Carcinoma: Current Status and Advances. Front Bioeng Biotechnol 2020; 8:184. [PMID: 32211399 PMCID: PMC7075945 DOI: 10.3389/fbioe.2020.00184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal solid cancers globally. To improve diagnosis sensitivities and treatment efficacies, the development of new theranostic nanoplatforms for efficient HCC management is urgently needed. In the past decade, mesoporous silica nanoparticles (MSNs) with tailored structure, large surface area, high agents loading volume, abundant chemistry functionality, acceptable biocompatibility have received more and more attention in HCC theranostic. This review outlines the recent advances in MSNs-based systems for HCC therapy and diagnosis. The multifunctional hybrid nanostructures that have both of therapy and diagnosis abilities are highlighted. And the precision delivery strategies of MSNs in HCC are also discussed. Final, we conclude with our personal perspectives on the future development and challenges of MSNs.
Collapse
Affiliation(s)
- Yaoye Tao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Chinese Academy of Medical Sciences (CAMS), Hangzhou, China
- Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Jianguo Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Chinese Academy of Medical Sciences (CAMS), Hangzhou, China
- Key Laboratory of Organ Transplantation, Hangzhou, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- National Health Commission (NHC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Chinese Academy of Medical Sciences (CAMS), Hangzhou, China
- Key Laboratory of Organ Transplantation, Hangzhou, China
| |
Collapse
|
19
|
Wu L, Zhang F, Chen X, Wan J, Wang Y, Li T, Wang H. Self-Assembled Gemcitabine Prodrug Nanoparticles Show Enhanced Efficacy against Patient-Derived Pancreatic Ductal Adenocarcinoma. ACS APPLIED MATERIALS & INTERFACES 2020; 12:3327-3340. [PMID: 31872760 DOI: 10.1021/acsami.9b16209] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Effective new therapies for pancreatic ductal adenocarcinoma (PDAC) are desperately needed as the prognosis of PDAC patients is dismal and treatment remains a major challenge. Gemcitabine (GEM) is commonly used to treat PDAC; however, the clinical use of GEM has been greatly compromised by its low delivery efficacy and drug resistance. Here, we describe a very simple yet cost-effective approach that synergistically combines drug reconstitution, supramolecular nanoassembly, and tumor-specific targeting to address the multiple challenges posed by the delivery of the chemotherapeutic drug GEM. Using our developed PUFAylation technology, the GEM prodrug was able to spontaneously self-assemble into colloidal stable nanoparticles with sub-100 nm size on covalent attachment of hydrophobic linoleic acid via amide linkage. The prodrug nanoassemblies could be further refined by PEGylation and PDAC-specific peptide ligand for preclinical studies. In vitro cell-based assays showed that not only were GEM nanoparticles superior to free GEM but also the decoration with PDAC-homing peptide facilitated the intracellular uptake of nanoparticles and thereby augmented the cytotoxic activity. In two separate xenograft models of human PDAC, one of which was a patient-derived xenograft model, the administration of targeted nanoparticles resulted in marked inhibition of tumor progression as well as alleviated systemic toxicity. Together, these data unequivocally confirm that the hydrophilic and rapidly metabolized drug GEM can be feasibly transformed into a pharmacologically efficient nanomedicine through exploiting the PUFAylation technology. This strategy could also potentially be applied to rescue many other therapeutics that show unfavorable outcomes in the preclinical studies because of pharmacologic obstacles.
Collapse
Affiliation(s)
- Liming Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| | - Fu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| | - Xiaona Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| | - Jianqin Wan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| | - Yuchen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
- Department of Chemical Engineering , Zhejiang University , Hangzhou 310027 , PR China
| | - Tongyu Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| | - Hangxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Zhejiang Provincial Key Laboratory of Pancreatic Disease; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine , Zhejiang University , Hangzhou 310003 , PR China
| |
Collapse
|
20
|
Han W, Shi L, Xie B, Wan J, Ren L, Wang Y, Chen X, Wang H. Supramolecular Engineering of Molecular Inhibitors in an Adaptive Cytotoxic Nanoparticle for Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:1707-1720. [PMID: 31816241 DOI: 10.1021/acsami.9b20178] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combinatorial regimens that rationally pair molecular inhibitors with standard cytotoxic chemotherapeutics are used to improve therapeutic outcomes. Simultaneously engineering these therapies within a single nanocarrier that spans cytotoxic, antiangiogenic, and anti-invasive mechanisms and that enables the delivery of unique drug combinations remains a technical challenge. In this study, we developed a simple and broadly applicable strategy in which ultrastable cytotoxic nanoparticles with an established excellent antitumor efficacy and π-rich inner core structure supramolecularly stabilized the antiangiogenic molecular inhibitor apatinib to create a synergistic drug delivery system (termed sTKI-pSN38). This small-sized nanoparticle accomplished the sequential release of both encapsulated drugs to exert antimetastatic, antivascular, and cytotoxic activities simultaneously. In xenograft models of hepatocellular carcinoma, a single intravenous administration of sTKI-pSN38 elicited robust and durable tumor reduction and suppressed metastasis to lymph nodes. Interestingly, sTKI-pSN38 treatment alleviated intratumoral hypoxia, which could contribute to impaired tumor metastasis and reduced drug resistance. Collectively, this nanotherapeutic platform offers a new strategy for cancer therapy by simply engineering a drug cocktail in conventional nanoparticles and by enabling the spatiotemporal modulation of drug release to enhance the synergy of the combined drugs.
Collapse
Affiliation(s)
- Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Linlin Shi
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Jianqin Wan
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Lulu Ren
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Yuchen Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Xiaona Chen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Hangxiang Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| |
Collapse
|
21
|
Dihydroartemisinin Sensitizes Mutant p53 (R248Q)-Expressing Hepatocellular Carcinoma Cells to Doxorubicin by Inhibiting P-gp Expression. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8207056. [PMID: 31976328 PMCID: PMC6955115 DOI: 10.1155/2019/8207056] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/04/2019] [Indexed: 02/07/2023]
Abstract
Mutant p53 (R248Q) induces doxorubicin (ADM) resistance in hepatocellular carcinoma (HCC). Dihydroartemisinin (DHA) can synergistically enhance anticancer effect of many chemotherapeutic agents. However, whether DHA could increase therapeutic efficacy of ADM in p53 (R248Q)-expressing HCC cells remains unknown. In the present study, we established mutant p53 (R248Q)-expressing Hep3B cells to study the effect and mechanism of DHA on ADM resistance and the synergistic effect of DHA with ADM. We found that P-gp was highly expressed in p53 (R248Q)-expressing Hep3B cells. As a result, cells expressing p53 (R248Q) displayed higher cell viability and lower cell apoptosis level upon ADM treatment. Meanwhile, phosphorylation levels of ERK1/2 and p65 were elevated in p53 (R248Q)-expressing Hep3B cells. However, combination of DHA and ADM treatment decreased cell viability and elevated cell apoptosis level in p53 (R248Q)-expressing Hep3B cells. Molecular dynamics simulations showed that DHA had the potential to bind with mutant p53 (R248Q) protein. Furthermore, DHA treatment decreased P-gp expression and inhibited phosphorylation levels of ERK1/2 and p65 in p53 (R248Q)-expressing Hep3B cells. Finally, DHA treatment could significantly reduce ADM efflux in p53 (R248Q)-expressing cells. Our results indicate that DHA could decrease P-gp expression via inhibiting the p53 (R248Q)-ERK1/2-NF-κB signaling pathway, which eventually confers sensitization of p53 (R248Q)-expressing HCC cells to ADM. Our study provides evidence for the potential application of DHA and ADM combination in treatment of mutant p53 (R248Q)-harbored HCC.
Collapse
|
22
|
Guo F, Fu Q, Jin C, Ji X, Yan Q, Yang Q, Wu D, Gao Y, Hong W, Li A, Yang G. Dual functional matrix metalloproteinase-responsive curcumin-loaded nanoparticles for tumor-targeted treatment. Drug Deliv 2019; 26:1027-1038. [PMID: 31691601 PMCID: PMC6844435 DOI: 10.1080/10717544.2019.1676843] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The limitations of anticancer drugs, including poor tumor targeting and weak uptake efficiency, are important factors affecting tumor therapy. According to characteristics of the tumor microenvironment, in this study, we aimed to synthesize matrix metalloproteinase (MMP)-responsive curcumin (Cur)-loaded nanoparticles (Cur-P-NPs) based on amphiphilic block copolymer (MePEG-peptide-PET-PCL) with MMP-cleavable peptide (GPLGIAGQ) and penetrating peptide (r9), modified to improve tumor targeting and cellular uptake. The average size of Cur-P-NPs was 176.9 nm, with a zeta potential of 8.1 mV, and they showed drug entrapment efficiency and a loading capacity of 87.07% ± 0.63% and 7.44% ± 0.16%, respectively. Furthermore, Cur release from Cur-P-NPs was sustained for 144 h at pH 7.4, and the release rate was accelerated under enzyme reaction condition. The MTT assay demonstrated that free P-NPs had favorable biosafety, and the anti-proliferative activity of Cur-P-NPs was positively correlated with Cur concentration in MCF-7 cells. Additionally, the results of cellular uptake, in vivo pharmacokinetics, and biodistribution showed that Cur-P-NPs had a good effect on cellular uptake and tumor targeting, resulting in the best bioavailability in tumor therapy. Therefore, Cur-P-NPs, as a promising drug delivery system, might lead to a new and efficient route for targeted therapy in clinical practice.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Qiafan Fu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Chenhao Jin
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Xugang Ji
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Qinying Yan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Qingliang Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Danjun Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Ying Gao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Weiyong Hong
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.,Taizhou Municipal Hospital of Zhejiang Province, Taizhou, China
| | - Aiqin Li
- Zhejiang Share Bio-pharm Co. Ltd, Hangzhou, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
23
|
Cheng G, Zhang X, Chen Y, Lee RJ, Wang J, Yao J, Zhang Y, Zhang C, Wang K, Yu B. Anticancer activity of polymeric nanoparticles containing linoleic acid-SN38 (LA-SN38) conjugate in a murine model of colorectal cancer. Colloids Surf B Biointerfaces 2019; 181:822-829. [DOI: 10.1016/j.colsurfb.2019.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 01/26/2023]
|
24
|
Nanomedicine as a putative approach for active targeting of hepatocellular carcinoma. Semin Cancer Biol 2019; 69:91-99. [PMID: 31421265 DOI: 10.1016/j.semcancer.2019.08.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/04/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
The effectiveness of chemotherapy in hepatocellular carcinoma (HCC) is restricted by chemo-resistance and systemic side effects. To improve the efficacy and safety of chemotherapeutics in HCC management, scientists have attempted to deliver these drugs to malignant tissues using targeted carriers as nanoparticles (NPs). Among the three types of NPs targeting (active, passive, and stimuli-responsive), active targeting is the most commonly investigated in HCC treatment. Despite the observed promising results so far, clinical research on nanomedicine targeting for HCC treatment still faces many challenges.These include batch-to-batch physicochemical properties' variations, limiting large scale production and insufficient data on human and environmental toxicities. This review summarized the characteristics of different nanocarriers, ligands, targeted receptors on HCC cells and provided recommendations to overcome the challenges, facing this novel line of treatment for HCC.
Collapse
|
25
|
Tahir N, Madni A, Correia A, Rehman M, Balasubramanian V, Khan MM, Santos HA. Lipid-polymer hybrid nanoparticles for controlled delivery of hydrophilic and lipophilic doxorubicin for breast cancer therapy. Int J Nanomedicine 2019; 14:4961-4974. [PMID: 31308666 PMCID: PMC6617603 DOI: 10.2147/ijn.s209325] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/14/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Lipid polymer hybrid nanoparticles (LPHNPs) for the controlled delivery of hydrophilic doxorubicin hydrochloride (DOX.HCl) and lipophilic DOX base have been fabricated by the single step modified nanoprecipitation method. Materials and methods: Poly (D, L-lactide-co-glicolide) (PLGA), lecithin, and 1,2-distearoyl-Sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000 (DSPE-PEG 2000) were selected as structural components. Results: The mean particle size was 173–208 nm, with an encapsulation efficiency of 17.8±1.9 to 43.8±4.4% and 40.3±0.6 to 59. 8±1.4% for DOX.HCl and DOX base, respectively. The drug release profile was in the range 33–57% in 24 hours and followed the Higuchi model (R2=0.9867–0.9450) and Fickian diffusion (n<0.5). However, the release of DOX base was slower than DOX.HCl. The in vitro cytotoxicity studies and confocal imaging showed safety, good biocompatibility, and a higher degree of particle internalization. The higher internalization of DOX base was attributed to higher permeability of lipophilic component and better hydrophobic interaction of particles with cell membranes. Compared to the free DOX, the DOX.HCl and DOX base loaded LPHNPs showed higher antiproliferation effects in MDA-MB231 and PC3 cells. Conclusion: Therefore, LPHNPs have provided a potential drug delivery strategy for safe, controlled delivery of both hydrophilic and lipophilic form of DOX in cancer cells.
Collapse
Affiliation(s)
- Nayab Tahir
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan.,Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.,Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Asadullah Madni
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Mubashar Rehman
- Department of Pharmacy, The University of central Pujnab, Lahore, Pakistan
| | - Vimalkumar Balasubramanian
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Muhammad Muzamil Khan
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland.,Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki FI-00014, Finland
| |
Collapse
|
26
|
Zhao M, Cui Y, Zhao L, Zhu T, Lee RJ, Liao W, Sun F, Li Y, Teng L. Thiophene Derivatives as New Anticancer Agents and Their Therapeutic Delivery Using Folate Receptor-Targeting Nanocarriers. ACS OMEGA 2019; 4:8874-8880. [PMID: 31459975 PMCID: PMC6648408 DOI: 10.1021/acsomega.9b00554] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/30/2019] [Indexed: 05/12/2023]
Abstract
A series of thiophene derivatives were synthesized by functionalization of 2,3-fused thiophene scaffolds. Their cytotoxicity was assessed against HeLa and Hep G2 cells. Compound 480 was identified as a promising candidate because of its low IC50 in HeLa (12.61 μg/mL) and Hep G2 (33.42 μg/mL) cells. The drug was loaded into folic acid (FA)-coated nanoparticles (NPs) to address its poor water solubility and to improve its selectivity for cancer cells. Compound 480 was shown to induce apoptosis by changes in mitochondrial membrane potential (ΔΨm) and the reactive oxygen species level. Furthermore, FA-modified NPs enhanced uptake capacity compared to unmodified controls by flow cytometry. This drug delivered in folate nanocarriers is promising for the treatment of cancers.
Collapse
Affiliation(s)
- Menghui Zhao
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Yaxin Cui
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Lang Zhao
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Tianyu Zhu
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Robert J. Lee
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
- College
of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus 43210, United States
| | - Weiwei Liao
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Fengying Sun
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Youxin Li
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
- E-mail: (Y.L.)
| | - Lesheng Teng
- School
of Life Sciences and College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
- E-mail: (L.T.)
| |
Collapse
|
27
|
Wu Y, Xu Z, Sun W, Yang Y, Jin H, Qiu L, Chen J, Chen J. Co-responsive smart cyclodextrin-gated mesoporous silica nanoparticles with ligand-receptor engagement for anti-cancer treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109831. [PMID: 31349481 DOI: 10.1016/j.msec.2019.109831] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/15/2019] [Accepted: 05/29/2019] [Indexed: 01/30/2023]
Abstract
Combination of both internal- and external-stimuli responsive strategies in nanoplatforms can maximize therapeutic outcomes by overcoming drug efflux-mediated resistance and prolonging sustained release of therapeutic payloads in controlled and sequential manner. Here, we show a light/redox dual-stimuli responsive β-cyclodextrin (β-CD)-gated mesoporous silica nanoparticles (MSN) that can effectively load and seal the chemotherapeutics, doxorubicin (DOX), inside MSN with a dual-capped system. The primary gatekeeper was achieved by capping β-CD via a disulfide linkage. An azobenzene/galactose-grafted polymer (GAP) was introduced to functionalize the MSN surface through host-guest interaction. GAP not only served as a secondary non-covalent polymer-gatekeeper to further prevent molecules from leaking out, but also presented targeting ligand for engagement of the asialoglycoprotein receptor (ASGPR) on hepatocellular carcinoma (HepG2) cells. The controlled and stimuli release of DOX could be realized via dissociation of azobenzene moieties from β-CD cage upon UV-irradiation, followed by liberation with the endogenous glutathione. The in vitro studies verified the redox-sensitive DOX release behavior, and the UV irradiation could accelerate this process to trigger DOX burst from MSN-ss-CD/GAP. Notably, the DOX@MSN-ss-CD/GAP could more efficiently deliver DOX into HepG2 cells and demonstrate enhanced cytotoxicity as compared with HeLa and COS7 cells. The smart MSN-ss-CD/GAP delivery system holds the potential for universal therapeutic uses in both biomedical research and clinical settings.
Collapse
Affiliation(s)
- Yaling Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Zheng Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Wenjing Sun
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Yingyue Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Hui Jin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Lipeng Qiu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China
| | - Jinghua Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China.
| | - Jingxiao Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
28
|
Yao X, Liu R, Liang X, Ding J. Critical Areas of Proliferation of Single Cells on Micropatterned Surfaces and Corresponding Cell Type Dependence. ACS APPLIED MATERIALS & INTERFACES 2019; 11:15366-15380. [PMID: 30964630 DOI: 10.1021/acsami.9b03780] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Material cues to influence cell proliferation are a fundamental issue in the fields of biomaterials, cell biology, tissue engineering, and regenerative medicine. This paper aims to investigate the proliferation of single mammal cells on micropatterned material surfaces. To this end, we prepared cell-adhesive circular microislands with 20 areas on the nonfouling background and systematically examined adhesion and proliferation behaviors of different kinds of single cells (primary stem and nonstem cells, cancer and normal cell lines) on micropatterns. On the basis of the analysis of experimental data, we found two critical areas about cell proliferation: (1) the critical spreading area of cells from almost no proliferation to confined proliferation, denoted as AP and (2) the critical spreading area of cells from confined proliferation to almost free proliferation, denoted as AFP. We further summarized the relative size relationship between these two critical areas and the characteristic areas of cell adhesion on both patterned and nonpatterned surfaces. While proliferation of single primary cells was affected by cell spreading, those cell lines, irrespective of normal and cancer cells, did not exhibit significant cell-spreading effects. As a result, this study reveals that proliferation of single cells is dependent upon spreading area, in particular for primary cells on material surfaces.
Collapse
Affiliation(s)
- Xiang Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , People's Republic of China
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , People's Republic of China
| | - Xiangyu Liang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , People's Republic of China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , People's Republic of China
| |
Collapse
|
29
|
Zhao Y, Lee RJ, Liu L, Dong S, Zhang J, Zhang Y, Yao Y, Lu J, Meng Q, Xie J, Teng L. Multifunctional drug carrier based on PEI derivatives loaded with small interfering RNA for therapy of liver cancer. Int J Pharm 2019; 564:214-224. [PMID: 31004717 DOI: 10.1016/j.ijpharm.2019.04.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/18/2019] [Accepted: 04/16/2019] [Indexed: 02/08/2023]
Abstract
Gene therapy strategies for liver cancer have broad application prospects but still lack a stable and efficient delivery vehicle. To overcome this obstacle, we designed a multifunctional gene delivery vector, sTPssOLP, which was based on oleylamine (OA)-modified disulfide-containing polyethylenimine (PEI) and incorporated into lipids to prepare a lipid nanoparticle. sTPssOLP consisted of the core of PEI derivative and cationic lipids bound to siRNA. The modified polyethylene glycol (PEG) and transferrin (Tf) were partially embedded in the phospholipid bilayer through the lipid and the other as the outer shell. The aim was to use the redox responsiveness of disulfide to trigger siRNA release in cytoplasm to enhance transfection efficiency. Pegylated lipids and Tf focus on increasing cycle life in the body and increasing accumulation at the tumor site of the carrier. In addition, two vectors were prepared as controls, one based on a PEI derivative containing no disulfide bond (POLP) and the other on the surface of the carrier not linked to Tf (PssOLP). PEI derivatives effectively avoid the toxicity problems caused by the use of PEI alone (25 kDa). Meanwhile, it was confirmed by gel retardation experiments that in the presence of dithiothreitol (DTT), the disulfide bond can indeed be reduced and the siRNA entrapped in the vector can be released. Both HepG2 and SMMC had significant uptake of sTPssOLP. The results of intracellular and lysosomal co-localization indicated that sTPssOLP achieved lysosomal escape. RT-PCR and Western blot results also confirmed that sTPssOLP had the best gene silencing activity. In vivo, the tumor inhibition rate of sTPssOLP in nude mice carrying HepG2 xenografts was 56%, which was significantly greater than that of the saline control group. In vivo imaging results showed that fluorescently labeled siRNA loaded in sTPssOLP was able to deliver more to the tumor site. At the same time, it was observed that sTPssOLP did not show significant damage to normal tissues. Therefore, this multifunctional gene delivery vector warrants further investigation.
Collapse
Affiliation(s)
- Yarong Zhao
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Robert J Lee
- Jilin University, School of Life Sciences, Changchun, Jilin, China; Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Luotong Liu
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Shiyan Dong
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Jing Zhang
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Yu Zhang
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | | | - Jiahui Lu
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Qingfan Meng
- Jilin University, School of Life Sciences, Changchun, Jilin, China
| | - Jing Xie
- Jilin University, School of Life Sciences, Changchun, Jilin, China.
| | - Lesheng Teng
- Jilin University, School of Life Sciences, Changchun, Jilin, China.
| |
Collapse
|
30
|
Li Y, Du L, Wu C, Yu B, Zhang H, An F. Peptide Sequence-Dominated Enzyme-Responsive Nanoplatform for Anticancer Drug Delivery. Curr Top Med Chem 2019; 19:74-97. [PMID: 30686257 DOI: 10.2174/1568026619666190125144621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
Enzymatic dysregulation in tumor and intracellular microenvironments has made this property
a tremendously promising responsive element for efficient diagnostics, carrier targeting, and drug
release. When combined with nanotechnology, enzyme-responsive drug delivery systems (DDSs) have
achieved substantial advancements. In the first part of this tutorial review, changes in tumor and intracellular
microenvironmental factors, particularly the enzymatic index, are described. Subsequently, the
peptide sequences of various enzyme-triggered nanomaterials are summarized for their uses in various
drug delivery applications. Then, some other enzyme responsive nanostructures are discussed. Finally,
the future opportunities and challenges are discussed. In brief, this review can provide inspiration and
impetus for exploiting more promising internal enzyme stimuli-responsive nanoDDSs for targeted tumor
diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Liping Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chunsheng Wu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
31
|
Jiang B, Yan L, Zhang J, Zhou M, Shi G, Tian X, Fan K, Hao C, Yan X. Biomineralization Synthesis of the Cobalt Nanozyme in SP94-Ferritin Nanocages for Prognostic Diagnosis of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:9747-9755. [PMID: 30777743 DOI: 10.1021/acsami.8b20942] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomaterials with intrinsic enzyme-like activities (nanozymes) have emerged as promising agents for cancer theranostics strategies. However, size-controllable synthesis of nanozymes and their targeting modifications are still challenging. Here, we report a monodispersed ferritin-based cobalt nanozyme (HccFn(Co3O4)) that specifically targets and visualizes clinical hepatocellular carcinoma (HCC) tissues. The cobalt nanozyme is biomimetically synthesized within the protein shell of the HCC-targeted ferritin (HccFn) nanocage, which is enabled by the display of HCC cell-specific peptide SP94 on the surface of ferritin through a genetic engineering approach. The intrinsic peroxidase-like activity of HccFn(Co3O4) nanozymes catalyzes the substrates to make color reaction to visualize HCC tumor tissues. We examined 424 clinical HCC specimens and verified that HccFn(Co3O4) nanozymes distinguish HCC tissues from normal liver tissues with a sensitivity of 63.5% and specificity of 79.1%, which is comparable with that of the clinically used HCC-specific marker α fetoprotein. Moreover, the pathological analysis indicates that the HccFn(Co3O4) nanozyme staining result is a potential predictor of prognosis in HCC patients. Staining intensity is positively correlated to tumor differentiation degree ( P = 0.0246) and tumor invasion ( P < 0.0001) and negatively correlated with overall survival ( P = 0.0084) of HCC patients. Together, our study demonstrates that ferritin is an excellent platform for size-controllable synthesis and targeting modifications of nanozymes, and the HccFn(Co3O4) nanozyme is a promising reagent for prognostic diagnosis of HCC.
Collapse
Affiliation(s)
- Bing Jiang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
- College of Life Sciences , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Liang Yan
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Jianlin Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Meng Zhou
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Guizhi Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Kelong Fan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
- College of Life Sciences , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
32
|
Wang Q, Zhang P, Li Z, Feng X, Lv C, Zhang H, Xiao H, Ding J, Chen X. Evaluation of Polymer Nanoformulations in Hepatoma Therapy by Established Rodent Models. Theranostics 2019; 9:1426-1452. [PMID: 30867842 PMCID: PMC6401493 DOI: 10.7150/thno.31683] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 01/10/2023] Open
Abstract
Hepatoma is one of the most severe malignancies usually with poor prognosis, and many patients are insensitive to the existing therapeutic agents, including the drugs for chemotherapy and molecular targeted therapy. Currently, researchers are committed to developing the advanced formulations with controlled drug delivery to improve the efficacy of hepatoma therapy. Numerous inoculated, induced, and genetically engineered hepatoma rodent models are now available for formulation screening. However, animal models of hepatoma cannot accurately represent human hepatoma in terms of histological characteristics, metastatic pathways, and post-treatment responses. Therefore, advanced animal hepatoma models with comparable pathogenesis and pathological features are in urgent need in the further studies. Moreover, the development of nanomedicines has renewed hope for chemotherapy and molecular targeted therapy of advanced hepatoma. As one kind of advanced formulations, the polymer-based nanoformulated drugs have many advantages over the traditional ones, such as improved tumor selectivity and treatment efficacy, and reduced systemic side effects. In this article, the construction of rodent hepatoma model and much information about the current development of polymer nanomedicines were reviewed in order to provide a basis for the development of advanced formulations with clinical therapeutic potential for hepatoma.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Chengyue Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Huaiyu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| |
Collapse
|
33
|
Liu R, Yao X, Liu X, Ding J. Proliferation of Cells with Severe Nuclear Deformation on a Micropillar Array. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:284-299. [PMID: 30513205 DOI: 10.1021/acs.langmuir.8b03452] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cellular responses on a topographic surface are fundamental topics about interfaces and biology. Herein, a poly(lactide- co-glycolide) (PLGA) micropillar array was prepared and found to trigger significant self-deformation of cell nuclei. The time-dependent cell viability and thus cell proliferation was investigated. Despite significant nuclear deformation, all of the examined cell types (Hela, HepG2, MC3T3-E1, and NIH3T3) could survive and proliferate on the micropillar array yet exhibited different proliferation abilities. Compared to the corresponding groups on the smooth surface, the cell proliferation abilities on the micropillar array were decreased for Hela and MC3T3-E1 cells and did not change significantly for HepG2 and NIH3T3 cells. We also found that whether the proliferation ability changed was related to whether the nuclear sizes decreased in the micropillar array, and thus the size deformation of cell nuclei should, besides shape deformation, be taken into consideration in studies of cells on topological surfaces.
Collapse
Affiliation(s)
- Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiang Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiangnan Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| |
Collapse
|
34
|
Wang Y, Chen X, He D, Zhou Y, Qin L. Surface-Modified Nanoerythrocyte Loading DOX for Targeted Liver Cancer Chemotherapy. Mol Pharm 2018; 15:5728-5740. [DOI: 10.1021/acs.molpharmaceut.8b00881] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yuemin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Xiaomei Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Dahua He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| | - Yi Zhou
- The College of Pharmaceutics Science, Guangzhou Medical University, Guangzhou, Guangdong 510436, China
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China
| |
Collapse
|
35
|
Ye PJ, Huang C, Yang S, Gao P, Li ZP, Tang SY, Xiang Y, Liu YF, Chen YP, He DX, Yu CY. Facile fabrication of a novel hybrid nanoparticles by self-assembling based on pectin-doxorubicin conjugates for hepatocellular carcinoma therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S661-S670. [PMID: 30307317 DOI: 10.1080/21691401.2018.1505745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the greatest public health problems worldwide, and chemotherapy remains the major approach for the HCC treatment. Doxorubicin (DOX) is one of the anthracycline antibiotics but its clinical use is limited due to its severe cardiotoxicity. In this study, novel hybrid nanoparticles by self-assembling based on pectin-doxorubicin conjugates (PDC-NPs) were fabricated for HCC treatment. The stabilized structure of the PDC-NPs was characterized by methylene blue absorption, the size, zeta potential and the morphology, which was investigated by Zetasizer nanoparticle analyzer and transmission electron microscope (TEM), of nanoparticles. The PDC-NPs achieved a sustained and prolonged release ability, which was illustrated with in vitro drug release profiles, anti-cell proliferation study, cellular uptake assay and in vivo pharmacokinetics analysis. Biocompatibility of the PDC-NPs was assessed with bovine serum albumin (BSA) adsorption test, hemolysis activity examination and viability evaluation of human umbilical vein endothelial cells. Importantly, in vivo studies of the PDC-NPs, which were performed in the athymic BALB/c nude mice, demonstrated that the PDC-NPs significantly reduced the lethal side effect of DOX. Additionally, the H&E staining and serum biochemistry study further confirmed the excellent biological security of the PDC-NPs.
Collapse
Affiliation(s)
- Peng-Ju Ye
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China.,b Institute of Pharmacy & Pharmacology , University of South China , Hengyang , China
| | - Can Huang
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Sa Yang
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Pei Gao
- b Institute of Pharmacy & Pharmacology , University of South China , Hengyang , China
| | - Zhi-Ping Li
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Si-Yue Tang
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Ya Xiang
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Yu-Feng Liu
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Yu-Ping Chen
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Dong-Xiu He
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China
| | - Cui-Yun Yu
- a Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study , University of South China , Hengyang , China.,b Institute of Pharmacy & Pharmacology , University of South China , Hengyang , China
| |
Collapse
|
36
|
Abdelmoneem MA, Mahmoud M, Zaky A, Helmy MW, Sallam M, Fang JY, Elkhodairy KA, Elzoghby AO. Decorating protein nanospheres with lactoferrin enhances oral COX-2 inhibitor/herbal therapy of hepatocellular carcinoma. Nanomedicine (Lond) 2018; 13:2377-2395. [DOI: 10.2217/nnm-2018-0134] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Lactoferrin (LF)-targeted gliadin nanoparticles (GL-NPs) were developed for targeted oral therapy of hepatocellular carcinoma. Materials & methods: Celecoxib and diosmin were incorporated in the hydrophobic matrix of GL-NPs whose surface was decorated with LF by electrostatic interaction for binding to asialoglycoprotein receptors overexpressed by liver cancer cells. Results: Targeted GL-NPs showed enhanced cytotoxic activity and increased cellular uptake in liver tumor cells compared with nontargeted NPs. Moreover, they demonstrated superior in vivo antitumor effects including reduction in the expression levels of tumor biomarkers and induction of caspase-mediated apoptosis. Ex vivo imaging of isolated organs exhibited extensive accumulation of NPs in livers more than other organs. Conclusion: LF-targeted GL-NPs could be considered as an efficient nanoplatform for targeted oral drug delivery for liver cancer therapy.
Collapse
Affiliation(s)
- Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mazen Mahmoud
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Amira Zaky
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Maged W Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhur University, Damanhur 22511, Egypt
| | - Marwa Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Industry of Human Ecology & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Kweishan, Taoyuan 333, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Division of Engineering in Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technologies, Cambridge, MA 02139, USA
| |
Collapse
|
37
|
Liu R, Jiang Y, Hu X, Wu J, Jiang W, Jin G, Luan Y. A preclinical evaluation of cytarabine prodrug nanofibers assembled from cytarabine-lauric acid conjugate toward solid tumors. Int J Pharm 2018; 552:111-118. [PMID: 30268848 DOI: 10.1016/j.ijpharm.2018.09.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/30/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022]
Abstract
Cytarabine (Ara-C) has become cornerstones for the treatment of hatmatological malignancies for several decades; however, it still faces serious challenges in clinical applications due to its side effects such as hand foot syndrome (HFS) and stomatitis. Therefore, considerable researchers have devoted to looking for the new derivative with desirable activity and low toxicity. A new prodrug based on the conjugation of cytarabine with lauric acid (LA-Ara) was synthesized in our group, and it could self-assemble into nanofibers (NFs) in aqueous solution with high drug loading (57 wt%). The lauric acid moiety protects NH2 group of from the enzymatic attachment and simultaneously raises the lipophilicity of Ara-C, thus obviously prolongs its plasma half-life. The oil/water partition coefficient (lg P) and the permeability of cell membrane of LA-Ara were obviously increased compared with Ara-C. Furthermore, the in vitro gastrointestinal stability results indicated the prodrug was suitable to be administrated orally. In the current study, the in vitro cytotoxicity and in vivo anti breast cancer experimental results indicate LA-Ara markedly improved antitumor activity compared with free Ara-C. The favorable safety evaluations elucidated its potentiality for oral alternative treatment to Ara-C. Importantly, LA-Ara can effectively decrease the incidence of toxic effects (HFS and stomatitis) of Ara-C, thereby exhibiting favorable skin safety profile. Overall, these results indicated the LA-Ara would be an excellent candidate for further clinical investigation and simultaneously highlight the prospects of Ara-C prodrug strategies in solid tumors therapy.
Collapse
Affiliation(s)
- Ruiling Liu
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Yue Jiang
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Xu Hu
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Jilian Wu
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Wei Jiang
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China
| | - Guoxia Jin
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, PR China
| | - Yuxia Luan
- School of Pharmaceutical Science, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, 44 West Wenhua Road, Jinan 250012, PR China.
| |
Collapse
|
38
|
Qiao Y, Wan J, Zhou L, Ma W, Yang Y, Luo W, Yu Z, Wang H. Stimuli‐responsive nanotherapeutics for precision drug delivery and cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1527. [DOI: 10.1002/wnan.1527] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/16/2018] [Accepted: 03/23/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Yiting Qiao
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| | - Jianqin Wan
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
- Department of Chemical Engineering Zhejiang University Hangzhou P.R. China
| | - Liqian Zhou
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| | - Wen Ma
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Yuanyuan Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Weixuan Luo
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening Southern Medical University Guangzhou P.R. China
| | - Hangxiang Wang
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi‐Organ Transplantation, Ministry of Public Health, School of Medicine Zhejiang University Hangzhou P.R. China
| |
Collapse
|
39
|
Zhang X, Zong W, Cheng W, Han X. Codelivery of doxorubicin and sodium tanshinone IIA sulfonate using multicompartmentalized vesosomes to enhance synergism and prevent doxorubicin-induced cardiomyocyte apoptosis. J Mater Chem B 2018; 6:5243-5247. [DOI: 10.1039/c8tb01136b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Doxorubicin, one of the most effective antitumor drugs, causes serious adverse cardiac effects.
Collapse
Affiliation(s)
- Xunan Zhang
- State Key Laboratory of Urban Water Resource and Environment
- School of Chemical Engineering and Technology
- Harbin Institute of Technology
- China
| | - Wei Zong
- State Key Laboratory of Urban Water Resource and Environment
- School of Chemical Engineering and Technology
- Harbin Institute of Technology
- China
| | - Wenlong Cheng
- Department of Chemical Engineering
- Monash University
- Australia
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment
- School of Chemical Engineering and Technology
- Harbin Institute of Technology
- China
| |
Collapse
|