1
|
Cui X, He Z, Liang J, Wei M, Guo Z, Zhou Y, Qin Y, Deng Z. Dehydrocurvularin-loaded mPEG-PLGA nanoparticles for targeted breast cancer drug delivery: preparation, characterization, in vitro, and in vivo evaluation. J Drug Target 2024; 32:325-333. [PMID: 38269592 DOI: 10.1080/1061186x.2024.2309566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/09/2023] [Indexed: 01/26/2024]
Abstract
Dehydrocurvularin (DCV) is a promising lead compound for anti-cancer therapy. Unfortunately, the development of DCV-based drugs has been hampered by its poor solubility and bioavailability. Herein, we prepared a DCV-loaded mPEG-PLGA nanoparticles (DCV-NPs) with improved drug properties and therapeutic efficacy. The spherical and discrete particles of DCV-NPs had a uniform diameter of 101.8 ± 0.45 nm and negative zeta potential of -22.5 ± 1.12 mV (pH = 7.4), and its entrapment efficiency (EE) and drug loading (DL) were ∼53.28 ± 1.12 and 10.23 ± 0.30%, respectively. In vitro the release of DCV-NPs lasted for more than 120 h in a sustained-release pattern, its antiproliferation efficacy towards breast cancer cell lines (MCF-7, MDA-MB-231, and 4T1) was better than that of starting drug DCV, and it could be efficiently and rapidly internalised by breast cancer cells. In vivo DCV-NPs were gradually accumulated in tumour areas of mice and significantly suppressed tumour growth. In summary, loading water-insoluble DCV onto nanoparticles has the potential to be an effective agent for breast cancer therapy with injectable property and tumour targeting capacity.
Collapse
Affiliation(s)
- Xuewei Cui
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, China
| | - Zhong He
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Jianjia Liang
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, China
| | - Mulan Wei
- Department of Pathology, Yiling Hospital Yichang, Yichang, China
| | - Zhiyong Guo
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, China
| | - Yiqing Zhou
- School of Biotechnology and Food Engineering, Changshu Institute of Technology, Changshu, China
| | - Ye Qin
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, China
| |
Collapse
|
2
|
Zou J, Xing X, Teng C, Zhao Q, He W, Wu X, Xia Y. Cocrystal@protein-anchoring nanococktail for combinatorially treating multidrug-resistant cancer. Acta Pharm Sin B 2024; 14:4509-4525. [PMID: 39525582 PMCID: PMC11544293 DOI: 10.1016/j.apsb.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/17/2024] [Accepted: 07/10/2024] [Indexed: 11/16/2024] Open
Abstract
Multidrug resistance (MDR), the major mechanism by which various cancers develop specific resistance to therapeutic agents, has set up enormous obstacles to many forms of tumor chemotherapy. Traditional cocktail therapy administration, based on the combination of multiple drugs for anti-MDR chemotherapy, often suffers from inconsistent in vivo pharmacokinetic behaviors that cannot act synchronously on the lesions, leading to limited pharmacodynamic outcomes. Despite the emergence of nanomedicines, which has improved chemotherapeutic drugs' bioavailability and therapeutic effect on clinical application, these monotherapy-based nano-formulations still show poor progression in overcoming MDR. Herein, a "one stone and three birds" nanococktail integrated by a cocrystal@protein-anchoring strategy was purposed for triple-payload delivery, which paclitaxel-disulfiram cocrystal-like nanorods (NRs) were anchored with the basic protein drug Cytochrome c (Cyt C), followed by hyaluronic-acid modification. In particular, NRs were utilized as carrier-like particles to synchronously deliver biomacromolecule Cyt C into tumor cells and then promote cell apoptosis. Of note, on A549/Taxol drug-resistant tumor-bearing mice, the system with extraordinarily high encapsulation efficiency demonstrated prolonged in vivo circulation and increased tumor-targeting accumulation, significantly reversing tumor drug resistance and improving therapeutic efficacy. Our mechanistic study indicated that the system induced the apoptosis of Taxol-resistant tumor cells through the signal axis P-glycoprotein/Cyt C/caspase 3. Collectively, this nanococktail strategy offers a promising approach to improve the sensitivity of tumor cells to chemotherapeutic drugs and strengthen intractable drug-resistant oncotherapy.
Collapse
Affiliation(s)
- Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xuyang Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingling Zhao
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xuri Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yuanzheng Xia
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
3
|
Deshmukh R, Sethi P, Singh B, Shiekmydeen J, Salave S, Patel RJ, Ali N, Rashid S, Elossaily GM, Kumar A. Recent Review on Biological Barriers and Host-Material Interfaces in Precision Drug Delivery: Advancement in Biomaterial Engineering for Better Treatment Therapies. Pharmaceutics 2024; 16:1076. [PMID: 39204421 PMCID: PMC11360117 DOI: 10.3390/pharmaceutics16081076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Preclinical and clinical studies have demonstrated that precision therapy has a broad variety of treatment applications, making it an interesting research topic with exciting potential in numerous sectors. However, major obstacles, such as inefficient and unsafe delivery systems and severe side effects, have impeded the widespread use of precision medicine. The purpose of drug delivery systems (DDSs) is to regulate the time and place of drug release and action. They aid in enhancing the equilibrium between medicinal efficacy on target and hazardous side effects off target. One promising approach is biomaterial-assisted biotherapy, which takes advantage of biomaterials' special capabilities, such as high biocompatibility and bioactive characteristics. When administered via different routes, drug molecules deal with biological barriers; DDSs help them overcome these hurdles. With their adaptable features and ample packing capacity, biomaterial-based delivery systems allow for the targeted, localised, and prolonged release of medications. Additionally, they are being investigated more and more for the purpose of controlling the interface between the host tissue and implanted biomedical materials. This review discusses innovative nanoparticle designs for precision and non-personalised applications to improve precision therapies. We prioritised nanoparticle design trends that address heterogeneous delivery barriers, because we believe intelligent nanoparticle design can improve patient outcomes by enabling precision designs and improving general delivery efficacy. We additionally reviewed the most recent literature on biomaterials used in biotherapy and vaccine development, covering drug delivery, stem cell therapy, gene therapy, and other similar fields; we have also addressed the difficulties and future potential of biomaterial-assisted biotherapies.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India;
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula 244236, India;
| | - Bhupendra Singh
- School of Pharmacy, Graphic Era Hill University, Dehradun 248002, India;
- Department of Pharmacy, S.N. Medical College, Agra 282002, India
| | | | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India;
| | - Ravish J. Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Anand 388421, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Arun Kumar
- School of Pharmacy, Sharda University, Greater Noida 201310, India
| |
Collapse
|
4
|
Wang Z, Yang L. Natural-product-based, carrier-free, noncovalent nanoparticles for tumor chemo-photodynamic combination therapy. Pharmacol Res 2024; 203:107150. [PMID: 38521285 DOI: 10.1016/j.phrs.2024.107150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
Cancer, with its diversity, heterogeneity, and complexity, is a significant contributor to global morbidity, disability, and mortality, highlighting the necessity for transformative treatment approaches. Photodynamic therapy (PDT) has aroused continuous interest as a viable alternative to conventional cancer treatments that encounter drug resistance. Nanotechnology has brought new advances in medicine and has shown great potential in drug delivery and cancer treatment. For precise and efficient therapeutic utilization of such a tumor therapeutic approach with high spatiotemporal selectivity and minimal invasiveness, the carrier-free noncovalent nanoparticles (NPs) based on chemo-photodynamic combination therapy is essential. Utilizing natural products as the foundation for nanodrug development offers unparalleled advantages, including exceptional pharmacological activity, easy functionalization/modification, and well biocompatibility. The natural-product-based, carrier-free, noncovalent NPs revealed excellent synergistic anticancer activity in comparison with free photosensitizers and free bioactive natural products, representing an alternative and favorable combination therapeutic avenue to improve therapeutic efficacy. Herein, a comprehensive summary of current strategies and representative application examples of carrier-free noncovalent NPs in the past decade based on natural products (such as paclitaxel, 10-hydroxycamptothecin, doxorubicin, etoposide, combretastatin A4, epigallocatechin gallate, and curcumin) for tumor chemo-photodynamic combination therapy. We highlight the insightful design and synthesis of the smart carrier-free NPs that aim to enhance PDT efficacy. Meanwhile, we discuss the future challenges and potential opportunities associated with these NPs to provide new enlightenment, spur innovative ideas, and facilitate PDT-mediated clinical transformation.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, PR China; School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus, Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, PR China; Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
5
|
Hou DY, Zhang NY, Wang L, Lv MY, Li XP, Zhang P, Wang YZ, Shen L, Wu XH, Fu B, Guo PY, Wang ZQ, Cheng DB, Wang H, Xu W. Inducing mitochondriopathy-like damages by transformable nucleopeptide nanoparticles for targeted therapy of bladder cancer. Natl Sci Rev 2024; 11:nwae028. [PMID: 38425424 PMCID: PMC10903983 DOI: 10.1093/nsr/nwae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/02/2024] [Accepted: 01/19/2024] [Indexed: 03/02/2024] Open
Abstract
Mitochondriopathy inspired adenosine triphosphate (ATP) depletions have been recognized as a powerful way for controlling tumor growth. Nevertheless, selective sequestration or exhaustion of ATP under complex biological environments remains a prodigious challenge. Harnessing the advantages of in vivo self-assembled nanomaterials, we designed an Intracellular ATP Sequestration (IAS) system to specifically construct nanofibrous nanostructures on the surface of tumor nuclei with exposed ATP binding sites, leading to highly efficient suppression of bladder cancer by induction of mitochondriopathy-like damages. Briefly, the reported transformable nucleopeptide (NLS-FF-T) self-assembled into nuclear-targeted nanoparticles with ATP binding sites encapsulated inside under aqueous conditions. By interaction with KPNA2, the NLS-FF-T transformed into a nanofibrous-based ATP trapper on the surface of tumor nuclei, which prevented the production of intracellular energy. As a result, multiple bladder tumor cell lines (T24, EJ and RT-112) revealed that the half-maximal inhibitory concentration (IC50) of NLS-FF-T was reduced by approximately 4-fold when compared to NLS-T. Following intravenous administration, NLS-FF-T was found to be dose-dependently accumulated at the tumor site of T24 xenograft mice. More significantly, this IAS system exhibited an extremely antitumor efficacy according to the deterioration of T24 tumors and simultaneously prolonged the overall survival of T24 orthotopic xenograft mice. Together, our findings clearly demonstrated the therapeutic advantages of intracellular ATP sequestration-induced mitochondriopathy-like damages, which provides a potential treatment strategy for malignancies.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Ni-Yuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Lu Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Mei-Yu Lv
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Peng Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Yue-Ze Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering & Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
| | - Xiu-Hai Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Bo Fu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Peng-Yu Guo
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Zi-Qi Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| |
Collapse
|
6
|
Kang C, Ren X, Lee D, Ramesh R, Nimmo S, Yang-Hartwich Y, Kim D. Harnessing small extracellular vesicles for pro-oxidant delivery: novel approach for drug-sensitive and resistant cancer therapy. J Control Release 2024; 365:286-300. [PMID: 37992875 PMCID: PMC10872719 DOI: 10.1016/j.jconrel.2023.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
Multidrug resistance (MDR) is an inevitable clinical problem in chemotherapy due to the activation of abundant P-glycoprotein (P-gp) that can efflux drugs. Limitations of current cancer therapy highlight the need for the development of a comprehensive cancer treatment strategy, including drug-resistant cancers. Small extracellular vesicles (sEVs) possess significant potential in surmounting drug resistance as they can effectively evade the efflux mechanism and transport small molecules directly to MDR cancer cells. One mechanism mediating MDR in cancer cells is sustaining increased levels of reactive oxygen species (ROS) and maintenance of the redox balance with antioxidants, including glutathione (GSH). Herein, we developed GSH-depleting benzoyloxy dibenzyl carbonate (B2C)-encapsulated sEVs (BsEVs), which overcome the efflux system to exert highly potent anticancer activity against human MDR ovarian cancer cells (OVCAR-8/MDR) by depleting GSH to induce oxidative stress and, in turn, apoptotic cell death in both OVCAR-8/MDR and OVCAR-8 cancer cells. BsEVs restore drug responsiveness by inhibiting ATP production through the oxidation of nicotinamide adenine dinucleotide with hydrogen (NADH) and inducing mitochondrial dysfunction, leading to the dysfunction of efflux pumps responsible for drug resistance. In vivo studies showed that BsEV treatment significantly inhibited the growth of OVCAR-8/MDR and OVCAR-8 tumors. Additionally, OVCAR-8/MDR tumors showed a trend towards a greater sensitivity to BsEVs compared to OVCAR tumors. In summary, this study demonstrates that BsEVs hold tremendous potential for cancer treatment, especially against MDR cancer cells.
Collapse
Affiliation(s)
- Changsun Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Xiaoyu Ren
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Jeonju 54896, South Korea
| | - Rajagopal Ramesh
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Susan Nimmo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
7
|
Ashique S, Garg A, Hussain A, Farid A, Kumar P, Taghizadeh‐Hesary F. Nanodelivery systems: An efficient and target-specific approach for drug-resistant cancers. Cancer Med 2023; 12:18797-18825. [PMID: 37668041 PMCID: PMC10557914 DOI: 10.1002/cam4.6502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Cancer treatment is still a global health challenge. Nowadays, chemotherapy is widely applied for treating cancer and reducing its burden. However, its application might be in accordance with various adverse effects by exposing the healthy tissues and multidrug resistance (MDR), leading to disease relapse or metastasis. In addition, due to tumor heterogeneity and the varied pharmacokinetic features of prescribed drugs, combination therapy has only shown modestly improved results in MDR malignancies. Nanotechnology has been explored as a potential tool for cancer treatment, due to the efficiency of nanoparticles to function as a vehicle for drug delivery. METHODS With this viewpoint, functionalized nanosystems have been investigated as a potential strategy to overcome drug resistance. RESULTS This approach aims to improve the efficacy of anticancer medicines while decreasing their associated side effects through a range of mechanisms, such as bypassing drug efflux, controlling drug release, and disrupting metabolism. This review discusses the MDR mechanisms contributing to therapeutic failure, the most cutting-edge approaches used in nanomedicine to create and assess nanocarriers, and designed nanomedicine to counteract MDR with emphasis on recent developments, their potential, and limitations. CONCLUSIONS Studies have shown that nanoparticle-mediated drug delivery confers distinct benefits over traditional pharmaceuticals, including improved biocompatibility, stability, permeability, retention effect, and targeting capabilities.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of PharmaceuticsPandaveswar School of PharmacyPandaveswarIndia
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, PharmacyJabalpurIndia
| | - Afzal Hussain
- Department of Pharmaceutics, College of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Arshad Farid
- Gomal Center of Biochemistry and BiotechnologyGomal UniversityDera Ismail KhanPakistan
| | - Prashant Kumar
- Teerthanker Mahaveer College of PharmacyTeerthanker Mahaveer UniversityMoradabadIndia
- Department of Pharmaceutics, Amity Institute of PharmacyAmity University Madhya Pradesh (AUMP)GwaliorIndia
| | - Farzad Taghizadeh‐Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of MedicineIran University of Medical SciencesTehranIran
- Clinical Oncology DepartmentIran University of Medical SciencesTehranIran
| |
Collapse
|
8
|
Yun WS, Kim J, Lim DK, Kim DH, Jeon SI, Kim K. Recent Studies and Progress in the Intratumoral Administration of Nano-Sized Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2225. [PMID: 37570543 PMCID: PMC10421122 DOI: 10.3390/nano13152225] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.
Collapse
Affiliation(s)
- Wan Su Yun
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongrae Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Kwon Lim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hwee Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
9
|
Mancino C, Pasto A, De Rosa E, Dolcetti L, Rasponi M, McCulloch P, Taraballi F. Immunomodulatory biomimetic nanoparticles target articular cartilage trauma after systemic administration. Heliyon 2023; 9:e16640. [PMID: 37313169 PMCID: PMC10258364 DOI: 10.1016/j.heliyon.2023.e16640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is one of the leading causes of disability in developed countries and accounts for 12% of all osteoarthritis cases in the United States. After trauma, inflammatory cells (macrophages amongst others) are quickly recruited within the inflamed synovium and infiltrate the joint space, initiating dysregulation of cartilage tissue homeostasis. Current therapeutic strategies are ineffective, and PTOA remains an open clinical challenge. Here, the targeting potential of liposome-based nanoparticles (NPs) is evaluated in a PTOA mouse model, during the acute phase of inflammation, in both sexes. NPs are composed of biomimetic phospholipids or functionalized with macrophage membrane proteins. Intravenous administration of NPs in the acute phase of PTOA and advanced in vivo imaging techniques reveal preferential accumulation of NPs within the injured joint for up to 7 days post injury, in comparison to controls. Finally, imaging mass cytometry uncovers an extraordinary immunomodulatory effect of NPs that are capable of decreasing the amount of immune cells infiltrating the joint and conditioning their phenotype. Thus, biomimetic NPs could be a powerful theranostic tool for PTOA as their accumulation in injury sites allows their identification and they have an intrinsic immunomodulatory effect.
Collapse
Affiliation(s)
- Chiara Mancino
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Anna Pasto
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Enrica De Rosa
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Luigi Dolcetti
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Patrick McCulloch
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, USA
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
10
|
Wang X, Wang Y, Yu J, Qiu Q, Liao R, Zhang S, Luo C. Reduction-Hypersensitive Podophyllotoxin Prodrug Self-Assembled Nanoparticles for Cancer Treatment. Pharmaceutics 2023; 15:784. [PMID: 36986645 PMCID: PMC10058384 DOI: 10.3390/pharmaceutics15030784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Podophyllotoxin (PPT) has shown strong antitumor effects on various types of cancers. However, the non-specific toxicity and poor solubility severely limits its clinical transformation. In order to overcome the adverse properties of PPT and explore its clinical potential, three novel PTT-fluorene methanol prodrugs linked by different lengths of disulfide bonds were designed and synthesized. Interestingly, the lengths of the disulfide bond affected the drug release, cytotoxicity, pharmacokinetic characteristics, in vivo biodistribution and antitumor efficacy of prodrug NPs. To be more specific, all three PPT prodrugs could self-assemble into uniform nanoparticles (NPs) with high drug loading (>40%) via the one-step nano precipitation method, which not only avoids the use of surfactants and cosurfactants, but also reduces the systemic toxicity of PPT and increases the tolerated dose. Among the three prodrug NPs, FAP NPs containing α-disulfide bond showed the most sensitive tumor-specific response and fastest drug release rate, thus demonstrating the strongest in vitro cytotoxicity. In addition, three prodrug NPs showed prolonged blood circulation and higher tumor accumulation. Finally, FAP NPs demonstrated the strongest in vivo antitumor activity. Our work will advance the pace of podophyllotoxin towards clinical cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
11
|
Podophyllotoxin and its derivatives: Potential anticancer agents of natural origin in cancer chemotherapy. Biomed Pharmacother 2023; 158:114145. [PMID: 36586242 DOI: 10.1016/j.biopha.2022.114145] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
The use of plant secondary metabolites has gained considerable attention among clinicians in the prevention and treatment of cancer. A secondary metabolite isolated mainly from the roots and rhizomes of Podophyllum species (Berberidaceae) is aryltetralin lignan - podophyllotoxin (PTOX). The purpose of this review is to discuss the therapeutic properties of PTOX as an important anticancer compound of natural origin. The relevant information regarding the antitumor mechanisms of podophyllotoxin and its derivatives were collected and analyzed from scientific databases. The results of the analysis showed PTOX exhibits potent cytotoxic activity; however, it cannot be used in its pure form due to its toxicity and generation of many side effects. Therefore, it practically remains clinically unusable. Currently, high effort is focused on attempts to synthesize analogs of PTOX that have better properties for therapeutic use e.g. etoposide (VP-16), teniposide, etopophos. PTOX derivatives are used as anticancer drugs which are showing additional immunosuppressive, antiviral, antioxidant, hypolipemic, and anti-inflammatory effects. In this review, attention is paid to the high potential of the usefulness of in vitro cultures of P. peltatum which can be a valuable source of lignans, including PTOX. In conclusion, the preclinical pharmacological studies in vitro and in vivo confirm the anticancer and chemotherapeutic potential of PTOX and its derivatives. In the future, clinical studies on human subjects are needed to certify the antitumor effects and the anticancer mechanisms to be certified and analyzed in more detail and to validate the experimental pharmacological preclinical studies.
Collapse
|
12
|
Han X, Alu A, Liu H, Shi Y, Wei X, Cai L, Wei Y. Biomaterial-assisted biotherapy: A brief review of biomaterials used in drug delivery, vaccine development, gene therapy, and stem cell therapy. Bioact Mater 2022; 17:29-48. [PMID: 35386442 PMCID: PMC8958282 DOI: 10.1016/j.bioactmat.2022.01.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Biotherapy has recently become a hotspot research topic with encouraging prospects in various fields due to a wide range of treatments applications, as demonstrated in preclinical and clinical studies. However, the broad applications of biotherapy have been limited by critical challenges, including the lack of safe and efficient delivery systems and serious side effects. Due to the unique potentials of biomaterials, such as good biocompatibility and bioactive properties, biomaterial-assisted biotherapy has been demonstrated to be an attractive strategy. The biomaterial-based delivery systems possess sufficient packaging capacity and versatile functions, enabling a sustained and localized release of drugs at the target sites. Furthermore, the biomaterials can provide a niche with specific extracellular conditions for the proliferation, differentiation, attachment, and migration of stem cells, leading to tissue regeneration. In this review, the state-of-the-art studies on the applications of biomaterials in biotherapy, including drug delivery, vaccine development, gene therapy, and stem cell therapy, have been summarized. The challenges and an outlook of biomaterial-assisted biotherapies have also been discussed.
Collapse
Affiliation(s)
- Xuejiao Han
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Liu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lulu Cai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Li M, Zhao Y, Sun J, Chen H, Liu Z, Lin K, Ma P, Zhang W, Zhen Y, Zhang S, Zhang S. pH/reduction dual-responsive hyaluronic acid-podophyllotoxin prodrug micelles for tumor targeted delivery. Carbohydr Polym 2022; 288:119402. [PMID: 35450654 DOI: 10.1016/j.carbpol.2022.119402] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 11/15/2022]
Abstract
Polymer-based prodrug nanocarriers with tumor-targeting and controlled-release properties are in great demand for enhanced cancer treatment. Hyaluronic acid (HA), which has excellent biocompatibility and targeting ability for cluster determinant 44 (CD44), has been proposed for delivering drugs that have poor solubility and high toxicity. Herein, podophyllotoxin (PPT) was conjugated to HA via ester and disulfide linkages to construct a pH- and reduction-responsive prodrug (HA-S-S-PPT). The micelles self-assembled from HA-S-S-PPT prodrug efficiently accumulated at tumor site due to HA receptor-mediated endocytosis. HA-S-S-PPT micelles exhibited 33.1% higher cumulative release than HA-NH-CO-PPT micelles (sensitive only to pH) owing to their dual responsiveness to pH and reduction. HA-S-S-PPT micelles achieved excellent antitumor activity in vivo, with the tumor inhibition rate reaching 92%, significantly higher than that of HA-NH-CO-PPT micelles (65%), and negligible systemic toxicity. This controllable-targeting nanoparticle system provides a potential platform for clinical application of PPT.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China; Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Jiao Sun
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Huiying Chen
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Zhanbiao Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Kexin Lin
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Pengfei Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Wenjun Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China; School of Chemical Engineering, Dalian University of Technology, Panjin 124221, PR China
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| | - Shufen Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China.
| |
Collapse
|
14
|
Ahmed T, Liu FCF, Lu B, Lip H, Park E, Alradwan I, Liu JF, He C, Zetrini A, Zhang T, Ghavaminejad A, Rauth AM, Henderson JT, Wu XY. Advances in Nanomedicine Design: Multidisciplinary Strategies for Unmet Medical Needs. Mol Pharm 2022; 19:1722-1765. [PMID: 35587783 DOI: 10.1021/acs.molpharmaceut.2c00038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Globally, a rising burden of complex diseases takes a heavy toll on human lives and poses substantial clinical and economic challenges. This review covers nanomedicine and nanotechnology-enabled advanced drug delivery systems (DDS) designed to address various unmet medical needs. Key nanomedicine and DDSs, currently employed in the clinic to tackle some of these diseases, are discussed focusing on their versatility in diagnostics, anticancer therapy, and diabetes management. First-hand experiences from our own laboratory and the work of others are presented to provide insights into strategies to design and optimize nanomedicine- and nanotechnology-enabled DDS for enhancing therapeutic outcomes. Computational analysis is also briefly reviewed as a technology for rational design of controlled release DDS. Further explorations of DDS have illuminated the interplay of physiological barriers and their impact on DDS. It is demonstrated how such delivery systems can overcome these barriers for enhanced therapeutic efficacy and how new perspectives of next-generation DDS can be applied clinically.
Collapse
Affiliation(s)
- Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Fuh-Ching Franky Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Brian Lu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Elliya Park
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Ibrahim Alradwan
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Jackie Fule Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Chunsheng He
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Abdulmottaleb Zetrini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Tian Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Amin Ghavaminejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Jeffrey T Henderson
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
15
|
pH-sensitive hyaluronic acid-targeted prodrug micelles constructed via a one-step reaction for enhanced chemotherapy. Int J Biol Macromol 2022; 206:489-500. [PMID: 35240214 DOI: 10.1016/j.ijbiomac.2022.02.131] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/09/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022]
Abstract
Although many chemotherapy prodrugs have been developed for tumor therapy, non-targeted delivery, uncontrolled release and tedious construction procedure of prodrugs still limit their clinical application in tumor treatment. In this work, hyaluronic acid (HA) which has tumor-targeting ability was used to conjugate to antitumor drug podophyllotoxin (PPT) to construct a pH-sensitive prodrug named HA-CO-O-PPT just via a one-step esterification reaction. The HA-CO-O-PPT spontaneously assembled into nano spherical micelles in aqueous medium, which had outstanding serum stability and blood compatibility. The obtained prodrug micelles (named HP micelles) exhibited a pH-responsive drug release mode with cumulative release reaching 81.2% due to their dissociation in response to acid stimulus, and had a high cellular uptake efficiency beyond 97% owing to HA receptor-mediated targeting. Furthermore, it was found that the prodrug micelles showed excellent antitumor activities in vivo with the tumor inhibition ratio up to 85% and negligible systemic toxicity. Accordingly, the pH-responsive HP micelles constructed by a simple one-step reaction, could be a promising candidate as a chemotherapeutic agent for cancer therapy.
Collapse
|
16
|
Figueiredo P, Lepland A, Scodeller P, Fontana F, Torrieri G, Tiboni M, Shahbazi MA, Casettari L, Kostiainen MA, Hirvonen J, Teesalu T, Santos HA. Peptide-guided resiquimod-loaded lignin nanoparticles convert tumor-associated macrophages from M2 to M1 phenotype for enhanced chemotherapy. Acta Biomater 2021; 133:231-243. [PMID: 33011297 DOI: 10.1016/j.actbio.2020.09.038] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Nanomedicines represent innovative and promising alternative technologies to improve the therapeutic effects of different drugs for cancer ablation. Targeting M2-like tumor-associated macrophages (TAMs) has emerged as a favorable therapeutic approach to fight against cancer through the modulation of the tumor microenvironment. However, the immunomodulatory molecules used for this purpose present side effects upon systemic administration, which limits their clinical translation. Here, the biocompatible lignin polymer is used to prepare lignin nanoparticles (LNPs) that carry a dual agonist of the toll-like receptors TLR7/8 (resiquimod, R848). These LNPs are targeted to the CD206-positive M2-like TAMs using the "mUNO" peptide, in order to revert their pro-tumor phenotype into anti-tumor M1-like macrophages in the tumor microenvironment of an aggressive triple-negative in vivo model of breast cancer. Overall, we show that targeting the resiquimod (R848)-loaded LNPs to the M2-like macrophages, using very low doses of R848, induces a profound shift in the immune cells in the tumor microenvironment towards an anti-tumor immune state, by increasing the representation of M1-like macrophages, cytotoxic T cells, and activated dendritic cells. This effect consequently enhances the anticancer effect of the vinblastine (Vin) when co-administered with R848-loaded LNPs. STATEMENT OF SIGNIFICANCE: Lignin-based nanoparticles (LNPs) were successfully developed to target a potent TLR7/8 agonist (R848) of the tumor microenvironment (TME). This was achieved by targeting the mannose receptor (CD206) on the tumor supportive (M2-like) macrophages with the "mUNO" peptide, to reprogram them into an anti-tumor (M1-like) phenotype for enhanced chemotherapy. LNPs modified the biodistribution of the R848, and enhanced its accumulation and efficacy in shifting the immunological profile of the cells in the TME, which was not achieved by systemic administration of free R848. Moreover, a reduction in the tumor volumes was observed at lower equivalent doses of R848 compared with other studies. Therefore, the co-administration of R848@LNPs is a promising chemotherapeutic application in aggressive tumors, such as the triple-negative breast cancer.
Collapse
Affiliation(s)
- Patrícia Figueiredo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Anni Lepland
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Pablo Scodeller
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia.
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Giulia Torrieri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mattia Tiboni
- Department of Biomolecular Sciences, School of Pharmacy, University of Urbino Carlo Bo, Urbino, Italy
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 56184-45139 Zanjan, Iran
| | - Luca Casettari
- Department of Biomolecular Sciences, School of Pharmacy, University of Urbino Carlo Bo, Urbino, Italy
| | - Mauri A Kostiainen
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, FI-00076, Aalto, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, 93106, CA, USA; Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, 92037, CA, USA.
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
17
|
Fan HY, Zhu ZL, Xian HC, Wang HF, Chen BJ, Tang YJ, Tang YL, Liang XH. Insight Into the Molecular Mechanism of Podophyllotoxin Derivatives as Anticancer Drugs. Front Cell Dev Biol 2021; 9:709075. [PMID: 34447752 PMCID: PMC8383743 DOI: 10.3389/fcell.2021.709075] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/22/2021] [Indexed: 02/05/2023] Open
Abstract
Podophyllotoxin (PTOX) is a biologically active compound derived from the podophyllum plant, and both it and its derivatives possess excellent antitumor activity. The PTOX derivatives etoposide (VP-16) and teniposide (VM-26) have been approved by the U.S. Food and Drug Administration (FDA) for cancer treatment, but are far from perfect. Hence, numerous PTOX derivatives have been developed to address the major limitations of PTOX, such as systemic toxicity, drug resistance, and low bioavailability. Regarding their anticancer mechanism, extensive studies have revealed that PTOX derivatives can induce cell cycle G2/M arrest and DNA/RNA breaks by targeting tubulin and topoisomerase II, respectively. However, few studies are dedicated to exploring the interactions between PTOX derivatives and downstream cancer-related signaling pathways, which is reasonably important for gaining insight into the role of PTOX. This review provides a comprehensive analysis of the role of PTOX derivatives in the biological behavior of tumors and potential molecular signaling pathways, aiming to help researchers design and develop better PTOX derivatives.
Collapse
Affiliation(s)
- Hua-yang Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Zhuo-li Zhu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Hong-chun Xian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Hao-fan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Bing-jun Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Ya-ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin-hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
18
|
Yang G, Liu Y, Zhao CX. Quantitative comparison of different fluorescent dye-loaded nanoparticles. Colloids Surf B Biointerfaces 2021; 206:111923. [PMID: 34146992 DOI: 10.1016/j.colsurfb.2021.111923] [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: 05/05/2021] [Revised: 06/07/2021] [Accepted: 06/13/2021] [Indexed: 12/18/2022]
Abstract
Labeling nanoparticles with fluorescent dyes is a common approach to investigate their cell uptake and biodistribution, providing valuable information for the preclinical assessment of nanoparticles for drug delivery. However, the underlying assumption that the fluorescence intensity of dye-labeled nanoparticles correlates positively with the amount of nanoparticles taken up by cells might not be valid under some conditions, as it can be affected by many factors including dye dispersion, dye quenching, and material shading. Here we demonstrated that both nanoparticles with hydrophobic dyes encapsulated inside and nanoparticles with hydrophilic dyes conjugated on the particle surface suffer from different degrees of dye quenching, making it challenging for quantitative comparison of cell uptake of different nanoparticles. To address this challenge, we proposed a possible solution for direct comparative studies of dye-labeled nanoparticles. This work provides valuable information for designing and evaluating different nanoparticles for drug delivery applications.
Collapse
Affiliation(s)
- Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Queensland, Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Queensland, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
19
|
Nguyen A, Böttger R, Li SD. Recent trends in bioresponsive linker technologies of Prodrug-Based Self-Assembling nanomaterials. Biomaterials 2021; 275:120955. [PMID: 34130143 DOI: 10.1016/j.biomaterials.2021.120955] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Prodrugs are designed to improve pharmaceutical properties of potent compounds and represent a central approach in drug development. The success of the prodrug strategy relies on incorporation of a reversible linkage facilitating controlled release of the parent drug. While prodrug approaches enhance pharmacokinetic properties over their parent drug, they still face challenges in absorption, distribution, metabolism, elimination, and toxicity (ADMET). Conjugating a drug to a carrier molecule such as a polymer can create an amphiphile that self-assembles into nanoparticles. These nanoparticles display prolonged blood circulation and passive targeting ability. Furthermore, the drug release can be tailored using a variety of linkers between the parent drug and the carrier molecule. In this review, we introduce the concept of self-assembling prodrugs and summarize different approaches for controlling the drug release with a focus on the linker technology. We also summarize recent clinical trials, discuss the emerging challenges, and provide our perspective on the utility and future potential of this technology.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
20
|
Lai X, Geng X, Li M, Tang M, Liu Q, Yang M, Shen L, Zhu Y, Wang S. Glutathione-responsive PLGA nanocomplex for dual delivery of doxorubicin and curcumin to overcome tumor multidrug resistance. Nanomedicine (Lond) 2021; 16:1411-1427. [PMID: 34047204 DOI: 10.2217/nnm-2021-0100] [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] [Indexed: 01/09/2023] Open
Abstract
Aim: This work aims to develop an injectable nano-drug delivery system to overcome tumor multidrug resistance (MDR). Methods: A drug delivery nanoplatform based on PEGylated PLGA with glutathione (GSH) responsivity was constructed for dual delivery of doxorubicin and curcumin (termed DCNP), and its MDR reversal efficiency was studied in vitro and in vivo. Results: The DCNPs exhibited a rapid drug release profile under high GSH concentration and could enhance the cellular uptake and cytotoxicity of doxorubicin to MDR cancer cells. Moreover, the DCNPs showed better biocompatibility, longer blood circulation and enhanced antitumor efficiency compared with free drugs. Conclusion: The GSH-responsive nanocarrier is believed to be a promising candidate for overcoming tumor MDR.
Collapse
Affiliation(s)
- Xuandi Lai
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| | - Xinran Geng
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, PR China
| | - Mengqing Li
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| | - Mengxiong Tang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| | - Qiong Liu
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| | - Mengsu Yang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, PR China
| | - Yu Zhu
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| | - Shubin Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science & Technology Medical Center, Shenzhen 518036, PR China
| |
Collapse
|
21
|
Yang G, Liu Y, Hui Y, Tengjisi, Chen D, Weitz DA, Zhao C. Implications of Quenching‐to‐Dequenching Switch in Quantitative Cell Uptake and Biodistribution of Dye‐Labeled Nanoparticles. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Yue Hui
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Tengjisi
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Dong Chen
- Institute of Process Equipment College of Energy Engineering Zhejiang University Hangzhou China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou China
| | - David A. Weitz
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA USA
- Department of Physics Harvard University Cambridge MA USA
| | - Chun‐Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
22
|
Yang G, Liu Y, Hui Y, Tengjisi, Chen D, Weitz DA, Zhao C. Implications of Quenching‐to‐Dequenching Switch in Quantitative Cell Uptake and Biodistribution of Dye‐Labeled Nanoparticles. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/anie.202101730] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Yue Hui
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Tengjisi
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| | - Dong Chen
- Institute of Process Equipment College of Energy Engineering Zhejiang University Hangzhou China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou China
| | - David A. Weitz
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA USA
- Department of Physics Harvard University Cambridge MA USA
| | - Chun‐Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
23
|
Li S, Li X, Lu Y, Hou M, Xu Z, Li B. A thiol-responsive and self-immolative podophyllotoxin prodrug for cancer therapy. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
24
|
Mthimkhulu NP, Mosiane KS, Nweke EE, Balogun M, Fru P. Prospects of Delivering Natural Compounds by Polymer-Drug Conjugates in Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:1699-1713. [PMID: 33874874 DOI: 10.2174/1871520621666210419094623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/01/2021] [Accepted: 03/08/2021] [Indexed: 11/22/2022]
Abstract
Synthetic chemotherapeutics have played a crucial role in minimizing mostly palliative symptoms associated with cancer; however, they have also created other problems such as system toxicity due to a lack of specificity. This has led to the development of polymer-drug conjugates amongst other novel drug delivery systems. Most of the formulations designed using delivery systems consist of synthetic drugs and face issues such as drug resistance, which has already rendered drugs such as antibiotics ineffective. This is further exacerbated by toxicity due to long term use. Given these problems and the fact that conjugation of synthetic compounds to polymers has been relatively slow with no formulation on the market after a decade of extensive studies, the focus has shifted to using this platform with medicinal plant extracts to improve solubility, specificity and increase drug release of medicinal and herbal bioactives. In recent years, various plant extracts such as flavonoids, tannins and terpenoids have been studied extensively using this approach. The success of formulations developed using novel drug-delivery systems is highly dependent on the tumour microenvironment especially on the enhanced permeability and retention effect. As a result, the compromised lymphatic network and 'leaky' vasculature exhibited by tumour cells act as a guiding principle in the delivering of these formulations. This review focuses on the state of the polymer-drug conjugates and their exploration with natural compounds, the progress and difficulties thus far, and future directions concerning cancer treatment.
Collapse
Affiliation(s)
- Nompumelelo P Mthimkhulu
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Karabo S Mosiane
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Ekene E Nweke
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Mohammed Balogun
- Biopolymer Modification and Therapeutics Lab, Materials Science & Manufacturing, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria 0001. South Africa
| | - Pascaline Fru
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| |
Collapse
|
25
|
Li C, Wang Y, Zhang S, Zhang J, Wang F, Sun Y, Huang L, Bian W. pH and ROS sequentially responsive podophyllotoxin prodrug micelles with surface charge-switchable and self-amplification drug release for combating multidrug resistance cancer. Drug Deliv 2021; 28:680-691. [PMID: 33818237 PMCID: PMC8023596 DOI: 10.1080/10717544.2021.1905750] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Multidrug resistance (MDR) is one of the main reasons for tumor chemotherapy failure. Podophyllotoxin (PPT) has been reported that can suppress MDR cancer cell growth; however, effective delivery of PPT to MDR cancer cells is challenged by cascaded bio-barriers. To effectively deliver PPT to MDR cancer cells, a PPT polymeric prodrug micelle (PCDMA) with the charge-conversion capability and self-acceleration drug release function are fabricated, which is composed of a pH and reactive oxygen species (ROS) sequentially responsive PPT-polymeric prodrug and an ROS generation agent, cucurbitacin B (CuB). After reach to tumor tissue, the surface charge of PCDMA could rapidly reverse to positive in the tumor extracellular environment to promote cellular uptake. Subsequently, the PCDMA could be degraded to release PPT and CuB in response to an intracellular high ROS condition. The released CuB is competent for generating ROS, which in turn accelerates the release of PPT and CuB. Eventually, the released PPT could kill MDR cancer cells. The in vitro and in vivo studies demonstrated that PCDMA was effectively internalized by cancer cells and produces massive ROS intracellular, rapid release drug, and effectively overcame MDR compared with the control cells, due to the tumor-specific weakly acidic and ROS-rich environment. Our results suggest that the pH/ROS dual-responsive PCDMA micelles with surface charge-reversal and self-amplifying ROS-response drug release provide an excellent platform for potential MDR cancer treatment.
Collapse
Affiliation(s)
- Chao Li
- Department of Infectious Disease, Wuhu No. 1 People's Hospital, Wuhu, China
| | - Yifan Wang
- Department of Oncology, Yancheng No. 1 People's Hospital, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Shuo Zhang
- Department of Infectious Disease, Wuhu No. 1 People's Hospital, Wuhu, China
| | - Jiaojiao Zhang
- Department of Infectious Disease, Wuhu No. 1 People's Hospital, Wuhu, China
| | - Fang Wang
- Department of Infectious Disease, Wuhu No. 1 People's Hospital, Wuhu, China
| | - Yunhao Sun
- Department of Cardiothoracic Surgery, Yancheng No. 1 People's Hospital, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Lirong Huang
- Department of Cardiothoracic Surgery, Yancheng No. 1 People's Hospital, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Wen Bian
- Department of Cardiothoracic Surgery, Yancheng No. 1 People's Hospital, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| |
Collapse
|
26
|
Chen Y, Pan Y, Hu D, Peng J, Hao Y, Pan M, Yuan L, Yu Y, Qian Z. Recent progress in nanoformulations of cabazitaxel. Biomed Mater 2021; 16:032002. [PMID: 33545700 DOI: 10.1088/1748-605x/abe396] [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] [Indexed: 02/08/2023]
Abstract
The antitumor efficacy of various paclitaxel (PTX) and docetaxel (DTX) formulations in clinical applications is seriously affected by drug resistance. Cabazitaxel, a second-generation taxane, exhibits greater anticancer activity than paclitaxel and docetaxel and has low affinity for the P-glycoprotein (P-gp) efflux pump because of its structure. Therefore, cabazitaxel has the potential to overcome taxane resistance. However, owing to the high systemic toxicity and hydrophobicity of cabazitaxel and the instability of its commercial preparation, Jevtana®, the clinical use of cabazitaxel is restricted to patients with metastatic castration-resistant prostate cancer (mCRPC) who show progression after docetaxel-based chemotherapy. Nanomedicine is expected to overcome the limitations associated with cabazitaxel application and surmount taxane resistance. This review outlines the drug delivery systems of cabazitaxel published in recent years, summarizes the challenges faced in the development of cabazitaxel nanoformulations, and proposes strategies to overcome these challenges.
Collapse
Affiliation(s)
- Yu Chen
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Yue Pan
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Danrong Hu
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Jinrong Peng
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Ying Hao
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Meng Pan
- Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Liping Yuan
- Sichuan University, Sichuan University, Chengdu, 610065, CHINA
| | - Yongyang Yu
- Department of Gastrointestinal Surgery, Sichuan University West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, CHINA
| | - Zhiyong Qian
- West China Hospital West China Medical School, Sichuan University, Sichuan University, Chengdu, 610041, CHINA
| |
Collapse
|
27
|
Kumbhar PS, Sakate AM, Patil OB, Manjappa AS, Disouza JI. Podophyllotoxin-polyacrylic acid conjugate micelles: improved anticancer efficacy against multidrug-resistant breast cancer. J Egypt Natl Canc Inst 2020; 32:42. [PMID: 33191444 DOI: 10.1186/s43046-020-00053-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/14/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Podophyllotoxin (PPT) is a naturally occurring compound obtained from the roots of Podophyllum species, indicated for a variety of malignant tumors such as breast, lung, and liver tumors. This toxic polyphenol (PPT) exhibited significant activity against P-glycoprotein (P-gp) mediated multidrug-resistant (MDR) cancer cells. However, extremely poor water solubility, a narrow therapeutic window, and high toxicity have greatly restricted the clinical uses of PPT. Therefore, the present research was aimed to synthesize the water-soluble ester prodrug of PPT with polyacrylic acid (PAA), a water-soluble polymer by Steglich esterification reaction, and to screen it for assay, solubility, in vitro hemolysis, in vitro release, and in vitro anticancer activity. RESULTS The Fourier transform infrared (FTIR) and nuclear magnetic resonance (NMR) spectroscopy results revealed the successful synthesis of podophyllotoxin-polyacrylic acid conjugate (PPC). The assay and saturation solubility of the prodrug is found to be 64.01 ± 4.5% and 1.39 ± 0.05 mg/mL (PPT equivalent) respectively. The PPC showed CMC (critical micelle concentration) of 0.430 mg/mL in distilled water at room temperature. The PPC micelles showed a mean particle size of 215 ± 11 nm with polydispersity index (PDI) of 0.193 ± 0.006. Further, the transmission electron microscope (TEM) results confirmed the self-assembling character of PPC into micelles. The PPC caused significantly less hemolysis (18.6 ± 2.9%) than plain PPT solution. Also, it demonstrated significantly (p < 0.01) higher in vitro cytotoxicity against both sensitive as well as resistance human breast cancer cells (MCF-7 and MDA MB-231) after 48 h of treatment. CONCLUSION The obtained study results clearly revealed the notable in vitro anticancer activity of PPT following its esterification with PAA. However, further in vivo studies are needed to ascertain its efficacy against a variety of cancers.
Collapse
Affiliation(s)
- Popat S Kumbhar
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Panhala, Kolhapur, Maharashtra, 416113, India
| | - Asmita M Sakate
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Panhala, Kolhapur, Maharashtra, 416113, India
| | - Onkar B Patil
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Panhala, Kolhapur, Maharashtra, 416113, India
| | - Arehalli S Manjappa
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Panhala, Kolhapur, Maharashtra, 416113, India
| | - John I Disouza
- Department of Pharmaceutics, Tatyasaheb Kore College of Pharmacy, Warananagar, Panhala, Kolhapur, Maharashtra, 416113, India.
| |
Collapse
|
28
|
Martinelli C, Biglietti M. Nanotechnological approaches for counteracting multidrug resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:1003-1020. [PMID: 35582219 PMCID: PMC8992571 DOI: 10.20517/cdr.2020.47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022]
Abstract
Every year, cancer accounts for a vast portion of deaths worldwide. Established clinical protocols are based on chemotherapy, which, however, is not tumor-selective and produces a series of unbearable side effects in healthy tissues. As a consequence, multidrug resistance (MDR) can arise causing metastatic progression and disease relapse. Combination therapy has demonstrated limited responses in the treatment of MDR, mainly due to the different pharmacokinetic properties of administered drugs and to tumor heterogeneity, challenges that still need to be solved in a significant percentage of cancer patients. In this perspective, we briefly discuss the most relevant MDR mechanisms leading to therapy failure and we report the most advanced strategies adopted in the nanomedicine field for the design and evaluation of ad hoc nanocarriers. We present some emerging classes of nanocarriers developed to reverse MDR and discuss recent progress evidencing their limits and promises.
Collapse
|
29
|
Zhao W, Cong Y, Li HM, Li S, Shen Y, Qi Q, Zhang Y, Li YZ, Tang YJ. Challenges and potential for improving the druggability of podophyllotoxin-derived drugs in cancer chemotherapy. Nat Prod Rep 2020; 38:470-488. [PMID: 32895676 DOI: 10.1039/d0np00041h] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covering: up to 2020As a main bioactive component of the Chinese, Indian, and American Podophyllum species, the herbal medicine, podophyllotoxin (PTOX) exhibits broad spectrum pharmacological activity, such as superior antitumor activity and against multiple viruses. PTOX derivatives (PTOXs) could arrest the cell cycle, block the transitorily generated DNA/RNA breaks, and blunt the growth-stimulation by targeting topoisomerase II, tubulin, or insulin-like growth factor 1 receptor. Since 1983, etoposide (VP-16) is being used in frontline cancer therapy against various cancer types, such as small cell lung cancer and testicular cancer. Surprisingly, VP-16 (ClinicalTrials NTC04356690) was also redeveloped to treat the cytokine storm in coronavirus disease 2019 (COVID-19) in phase II in April 2020. The treatment aims at dampening the cytokine storm and is based on etoposide in the case of central nervous system. However, the initial version of PTOX was far from perfect. Almost all podophyllotoxin derivatives, including the FDA-approved drugs VP-16 and teniposide, were seriously limited in clinical therapy due to systemic toxicity, drug resistance, and low bioavailability. To meet this challenge, scientists have devoted continuous efforts to discover new candidate drugs and have developed drug strategies. This review focuses on the current clinical treatment of PTOXs and the prospective analysis for improving druggability in the rational design of new generation PTOX-derived drugs.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wang Q, Sun M, Li C, Li D, Yang Z, Jiang Q, He Z, Ding H, Sun J. A computer-aided chem-photodynamic drugs self-delivery system for synergistically enhanced cancer therapy. Asian J Pharm Sci 2020; 16:203-212. [PMID: 33995614 PMCID: PMC8105418 DOI: 10.1016/j.ajps.2020.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/28/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023] Open
Abstract
The therapeutic strategy that gives consideration to the combination of photodynamic therapy and chemotherapy, has emerged as a potential development of effective anti-cancer medicine. Nevertheless, co-delivery of photosensitizers (PSs) and chemotherapeutic drugs in traditional carriers still remains great limitations due to low drug loadings and poor biocompatibility. Herein, we have utilized a computer-aided strategy to achieve a desired carrier-free self-delivery of pyropheophorbide a (PPa, a common PS) and podophyllotoxin (PPT, a classical chemotherapeutic drug) for synergistic cancer therapy. First, the computational simulation method identified the similar molecular sizes and rigid molecular structures between two drugs molecules. Based on the molecular docking, the intermolecular interactions were found to include π-π stackings, hydrophobic interactions and hydrogen bonds. Next, both drugs could co-assemble into nanoparticles (NPs) via one-step nanoprecipitation method. The various spectral experiments (UV, IR and FL) were conducted to evaluate the formation mechanism of spherical NPs. Moreover, in vitro and in vivo experiments systematically demonstrated that PPT/PPa NPs not only showed better cellular uptake efficiency, stronger cytotoxicity and higher accumulation in tumor sites, but also exhibited synergistic antitumor effect in female BALB/C bearing-4T1 tumor mice. Such a computer-aided design strategy of chem-photodynamic drugs self-delivery systems pave the way for efficient synergistic cancer therapy.
Collapse
Affiliation(s)
- Qiu Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengchi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chang Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zimeng Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qikun Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
31
|
Costea T, Vlad OC, Miclea LC, Ganea C, Szöllősi J, Mocanu MM. Alleviation of Multidrug Resistance by Flavonoid and Non-Flavonoid Compounds in Breast, Lung, Colorectal and Prostate Cancer. Int J Mol Sci 2020; 21:E401. [PMID: 31936346 PMCID: PMC7013436 DOI: 10.3390/ijms21020401] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the manuscript is to discuss the influence of plant polyphenols in overcoming multidrug resistance in four types of solid cancers (breast, colorectal, lung and prostate cancer). Effective treatment requires the use of multiple toxic chemotherapeutic drugs with different properties and targets. However, a major cause of cancer treatment failure and metastasis is the development of multidrug resistance. Potential mechanisms of multidrug resistance include increase of drug efflux, drug inactivation, detoxification mechanisms, modification of drug target, inhibition of cell death, involvement of cancer stem cells, dysregulation of miRNAs activity, epigenetic variations, imbalance of DNA damage/repair processes, tumor heterogeneity, tumor microenvironment, epithelial to mesenchymal transition and modulation of reactive oxygen species. Taking into consideration that synthetic multidrug resistance agents have failed to demonstrate significant survival benefits in patients with different types of cancer, recent research have focused on beneficial effects of natural compounds. Several phenolic compounds (flavones, phenolcarboxylic acids, ellagitannins, stilbens, lignans, curcumin, etc.) act as chemopreventive agents due to their antioxidant capacity, inhibition of proliferation, survival, angiogenesis, and metastasis, modulation of immune and inflammatory responses or inactivation of pro-carcinogens. Moreover, preclinical and clinical studies revealed that these compounds prevent multidrug resistance in cancer by modulating different pathways. Additional research is needed regarding the role of phenolic compounds in the prevention of multidrug resistance in different types of cancer.
Collapse
Affiliation(s)
- Teodora Costea
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Oana Cezara Vlad
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - Luminita-Claudia Miclea
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Research Excellence Center in Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Constanta Ganea
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Maria-Magdalena Mocanu
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| |
Collapse
|
32
|
Feng W, Zong M, Wan L, Yu X, Yu W. pH/redox sequentially responsive nanoparticles with size shrinkage properties achieve deep tumor penetration and reversal of multidrug resistance. Biomater Sci 2020; 8:4767-4778. [PMID: 32724941 DOI: 10.1039/d0bm00695e] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
pH/redox sequentially responsive nanoparticles with size shrinkage properties achieve deep tumor penetration and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Wanting Feng
- Department of Oncology
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University
- Huaian
- China
| | - Mingzhu Zong
- Department of Oncology
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University
- Huaian
- China
| | - Li Wan
- Department of Oncology
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University
- Huaian
- China
| | - Xiaojuan Yu
- Department of Oncology
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University
- Huaian
- China
| | - Weiyong Yu
- Department of Oncology
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University
- Huaian
- China
| |
Collapse
|
33
|
Nguyen A, Ando H, Böttger R, DurgaRao Viswanadham KK, Rouhollahi E, Ishida T, Li SD. Utilization of click chemistry to study the effect of poly(ethylene)glycol molecular weight on the self-assembly of PEGylated gambogic acid nanoparticles for the treatment of rheumatoid arthritis. Biomater Sci 2020; 8:4626-4637. [DOI: 10.1039/d0bm00711k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Click chemistry was used to study the effect of varied PEG molecular weights on the self-assembly of PEG-gambogic acid (GA) conjugates into nanoparticles.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics
- Subdivision of Biopharmaceutical Sciences
- Institute of Health Biosciences
- The University of Tokushima
- Tokushima
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | | | - Elham Rouhollahi
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics
- Subdivision of Biopharmaceutical Sciences
- Institute of Health Biosciences
- The University of Tokushima
- Tokushima
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| |
Collapse
|
34
|
Kumari A, Srivastava S, Manne RK, Sisodiya S, Santra MK, Guchhait SK, Panda D. C12, a combretastatin-A4 analog, exerts anticancer activity by targeting microtubules. Biochem Pharmacol 2019; 170:113663. [PMID: 31606408 DOI: 10.1016/j.bcp.2019.113663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022]
Abstract
Combretastatin A4 and its analogs are undergoing various clinical trials for the treatment of different cancers. This study illustrated the molecular mechanism and antitumor activity of C12, (5-Quinolin-3-yl and 4-(3,4,5-trimethoxyphenyl) substituted imidazol-2-amine), a synthetic analog of CA-4. C12 reduced the tumor volume of MCF-7 xenograft in NOD-SCID mice without affecting the bodyweight of the mice. Further, C12 inhibited the proliferation of several types of cancer cells more efficiently than their noncancerous counterparts. Using GFP-EB1 imaging, the effects of C12 on the interphase microtubule dynamics were determined in live HeLa cells. C12 (10 nM, half-maximal proliferation inhibitory concentration) reduced the growth rate of microtubules by 52% and increased the pause time of microtubules by 68%. In addition, fluorescence recovery after photobleaching analysis demonstrated that 10 nM C12 strongly suppressed spindle microtubule dynamics in HeLa cells. C12 treatment reduced the interpolar distance between the two spindle poles, increased the chromosome congression index, inhibited chromosome movement, and increased the level of mitotic checkpoint complex proteins BubR1 and Mad2. The evidence presented here indicated that C12 could induce different modes of cell death, depending on the extent of microtubule depolymerization. Since C12 targets both the mitotic and non-mitotic cells and showed a stronger activity against cancerous cells than non-cancerous cells, it may have an advantage in cancer chemotherapy. The results significantly enhance our understanding of the antitumor mechanism of the microtubule-depolymerizing agents.
Collapse
Affiliation(s)
- Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shalini Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Rajesh K Manne
- National Centre for Cell Science, University of Pune Campus, Pune, Maharashtra 411007, India
| | - Shailendra Sisodiya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S. A. S. Nagar, Punjab 160062, India
| | - Manas K Santra
- National Centre for Cell Science, University of Pune Campus, Pune, Maharashtra 411007, India.
| | - Sankar K Guchhait
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S. A. S. Nagar, Punjab 160062, India.
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India.
| |
Collapse
|
35
|
Ag Seleci D, Maurer V, Stahl F, Scheper T, Garnweitner G. Rapid Microfluidic Preparation of Niosomes for Targeted Drug Delivery. Int J Mol Sci 2019; 20:ijms20194696. [PMID: 31546717 PMCID: PMC6801367 DOI: 10.3390/ijms20194696] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022] Open
Abstract
Niosomes are non-ionic surfactant-based vesicles with high promise for drug delivery applications. They can be rapidly prepared via microfluidics, allowing their reproducible production without the need of a subsequent size reduction step, by controlled mixing of two miscible phases of an organic (lipids dissolved in alcohol) and an aqueous solution in a microchannel. The control of niosome properties and the implementation of more complex functions, however, thus far are largely unknown for this method. Here we investigate microfluidics-based manufacturing of topotecan (TPT)-loaded polyethylene glycolated niosomes (PEGNIO). The flow rate ratio of the organic and aqueous phases was varied and optimized. Furthermore, the surface of TPT-loaded PEGNIO was modified with a tumor homing and penetrating peptide (tLyp-1). The designed nanoparticular drug delivery system composed of PEGNIO-TPT-tLyp-1 was fabricated for the first time via microfluidics in this study. The physicochemical properties were determined through dynamic light scattering (DLS) and zeta potential analysis. In vitro studies of the obtained formulations were performed on human glioblastoma (U87) cells. The results clearly indicated that tLyp-1-functionalized TPT-loaded niosomes could significantly improve anti-glioma treatment.
Collapse
Affiliation(s)
- Didem Ag Seleci
- Institute for Particle Technology (iPAT), Technische Universität Braunschweig, 38104 Braunschweig, Germany.
- Centre for Pharmaceutical Engineering Research (PVZ), Technische Universität Braunschweig, 38106 Braunschweig, Germany.
| | - Viktor Maurer
- Institute for Particle Technology (iPAT), Technische Universität Braunschweig, 38104 Braunschweig, Germany.
- Centre for Pharmaceutical Engineering Research (PVZ), Technische Universität Braunschweig, 38106 Braunschweig, Germany.
| | - Frank Stahl
- Institute for Technical Chemistry, Leibniz University Hannover, 30167 Hannover, Germany.
| | - Thomas Scheper
- Institute for Technical Chemistry, Leibniz University Hannover, 30167 Hannover, Germany.
| | - Georg Garnweitner
- Institute for Particle Technology (iPAT), Technische Universität Braunschweig, 38104 Braunschweig, Germany.
- Centre for Pharmaceutical Engineering Research (PVZ), Technische Universität Braunschweig, 38106 Braunschweig, Germany.
| |
Collapse
|
36
|
Dai L, Si C. Recent Advances on Cellulose-Based Nano-Drug Delivery Systems: Design of Prodrugs and Nanoparticles. Curr Med Chem 2019; 26:2410-2429. [PMID: 28699504 DOI: 10.2174/0929867324666170711131353] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/10/2017] [Accepted: 03/31/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cellulose being the first abundant biopolymers in nature has many fascinating properties, including low-cost, good biodegradability, and excellent biocompatibility, which made cellulose a real potential material to create nano-drug delivery systems (nano-DDS). This review aims to present and discuss some remarkable recent advances on the drug delivery applications of cellulosebased prodrugs and nanoparticles. METHODS By searching the research literatures over last decade, a variety featured studies on cellulosebased nano-DDS were summarized and divided into prodrugs, prodrug nanoparticles, solid or derivative nanopartilces, amphiphilic copolymer nanoparticles, and polyelectrolyte complex nanoparticles. Various methods employed for the functionalization, pharmacodynamic actions and applications were described and discussed. RESULTS Many types of cellulose-based nano-DDS can ensure efficient encapsulation of various drugs and then overcome the free drug molecule shortcomings. Among all the method described, cellulosebased amphiphilic nanoparticles are most frequently used. These formulations have the higher drug loading capability, a simple and flexible way to achieve multi-functional. Apart from hydrophilic or hydrophobic modification, cellulose or its derivatives can form nanoparticles with different small molecules and macromolecules, leading to a large spectrum of cellulose-based nano-DDS and providing some unexpected advantages. CONCLUSION Thorough physicochemical characterization and profound understanding of interactions of the cellulose-based nano-DDS with cells and tissues is indispensable. Moreover, studies toward technics parameter optimization and scale up from the laboratory to production level should be undertaken. The development of intravenous and orally applicable cellulose-based nano-DDS will be an important research area, and these systems will have more commercial status in the market.
Collapse
Affiliation(s)
- Lin Dai
- Tianjin Key Laboratory of Pulp and Paper, Tianjin University of Science and Technology, Tianjin 300457, China.,State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China
| | - Chuanling Si
- Tianjin Key Laboratory of Pulp and Paper, Tianjin University of Science and Technology, Tianjin 300457, China.,State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China.,State Key Laboratory of Tree Genetics and Breeding, Chinese Academy of Forestry, Beijing 100091, China
| |
Collapse
|
37
|
Li Y, Chen M, Yao B, Lu X, Zhang X, He P, Vasilatos SN, Ren X, Bian W, Yao C. Transferrin receptor-targeted redox/pH-sensitive podophyllotoxin prodrug micelles for multidrug-resistant breast cancer therapy. J Mater Chem B 2019; 7:5814-5824. [PMID: 31495855 DOI: 10.1039/c9tb00651f] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Podophyllotoxin (PPT), a toxic polyphenol extracted from the roots of Podophyllum species, showed remarkable activity against P-glycoprotein (P-gp) mediated multidrug resistant (MDR) cancer cells. Many PPT-prodrugs based on nano-technology have been developed for increasing aqueous solubility and reducing the side effects of PPT; however, the sensitive linkers in almost all PPT-prodrugs were ester bonds, resulting in slow and incomplete drug release. We developed a redox/pH double-sensitive and tumor active targeted drug delivery system for PPT delivery, in which PPT was covalently coupled to T7-peptide (Pep) modified polyethylene glycol (PEG) or methoxy-polyethylene glycol (mPEG) through a disulfide bond to obtain the final polymer (Pep-PEG-SS-PPT or PEG-SS-PPT). The mixed micelles (Pep-SS-NPs) were made by mixing Pep-PEG-SS-PPT with PEG-SS-PPT, and the mixed micelles showed good size uniformity and high stability in serum solution. The in vitro release experiment showed that about (81.7 ± 2.8)% PPT was released from Pep-SS-NPs in 10 mM glutathione (GSH) at pH 7.4, and also about (64.6 ± 1.7)% PPT was released from Pep-SS-NPs at pH 5.0. In vitro cytotoxicity analysis suggested that Pep-SS-NPs exhibited 57- to 270-fold lower resistance index (RI) values for different drug-resistant cancer cell lines than paclitaxel (PTX) or docetaxel (DTX). The cell uptake assay indicated that the Pep-SS-NPs could significantly enhance the intracellular level of coumarin-6 compared to that of the control group. The maximum tolerated dose (MTD) of Pep-SS-NPs was increased greatly compared to that of free PPT (5.3-fold). In vivo research showed that Pep-SS-NPs significantly enhanced antitumor efficacy against MCF-7/ADR xenograft tumors compared to the control groups. These findings suggest that mixed micelles could be a potentially successful nanomedicine for MDR breast cancer therapy.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| | - Mie Chen
- Department of general surgery, Pukou district central hospital, Pukou branch of jiangsu province hospital, China
| | - Bowen Yao
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xun Lu
- Milken School of Public Health, George Washington University, Washington, DC 20052, USA
| | - Xiaoqing Zhang
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| | - Peng He
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| | - Shauna N Vasilatos
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaomei Ren
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| | - Weihe Bian
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| | - Chang Yao
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), No. 155 Hanzhong Road, Nanjing 210029, China.
| |
Collapse
|
38
|
Li F, Liu D, Liao X, Zhao Y, Li R, Yang B. Acid-controlled release complexes of podophyllotoxin and etoposide with acyclic cucurbit[n]urils for low cytotoxicity. Bioorg Med Chem 2019; 27:525-532. [DOI: 10.1016/j.bmc.2018.12.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/14/2018] [Accepted: 12/27/2018] [Indexed: 02/02/2023]
|
39
|
Tumor heterogeneity and nanoparticle-mediated tumor targeting: the importance of delivery system personalization. Drug Deliv Transl Res 2018; 8:1508-1526. [PMID: 30128797 DOI: 10.1007/s13346-018-0578-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
After the discovery of the enhanced permeability and retention effect in 1986, it was envisioned that nanoparticle-mediated tumor-targeted delivery of chemotherapeutics would make a radical change in cancer therapy. However, after three decades of extensive research, only a few nanotherapeutics have been approved for clinical use. Although significant advantages of nanomedicines have been demonstrated in pre-clinical studies, clinical outcome was found to be variable. Advanced research has revealed that significant biochemical and structural variations exist between (and among) different tumors. These variations can considerably affect the tumor delivery and efficacy of nanomedicines. Tumor penetration is an important determining factor for positive therapeutic outcome and same nanomedicine can show diverse efficacy against different tumors depending on the extent of tumor accumulation and penetration. Recent research has started shading light on how the tumor variations can influence nanoparticle tumor delivery. These findings indicate that there is no "ideal" design of nanoparticles for exhibiting equally high efficacy against a broad spectrum of tumors. For achieving maximum benefit of the nanotherapeutics, it is necessary to analyze the tumor microenvironment for understanding the biological and structural characteristics of the tumor. Designing of the nanomedicine should be done according to the tumor characteristics. In this comprehensive review, we have first given a brief overview of the design characteristics of nanomedicine which impact their tumor delivery. Then we discussed about the variability in the tumor architecture and how it influences nanomedicine delivery. Finally, we have discussed the possibility of delivery system personalization based on the tumor characteristics.
Collapse
|
40
|
Bumbaca B, Li W. Taxane resistance in castration-resistant prostate cancer: mechanisms and therapeutic strategies. Acta Pharm Sin B 2018; 8:518-529. [PMID: 30109177 PMCID: PMC6089846 DOI: 10.1016/j.apsb.2018.04.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Despite its good initial response and significant survival benefit in patients with castration-resistant prostate cancer (CRPC), taxane therapy inevitably encounters drug resistance in all patients. Deep understandings of taxane resistant mechanisms can significantly facilitate the development of new therapeutic strategies to overcome taxane resistance and improve CRPC patient survival. Multiple pathways of resistance have been identified as potentially crucial areas of intervention. First, taxane resistant tumor cells typically have mutated microtubule binding sites, varying tubulin isotype expression, and upregulation of efflux transporters. These mechanisms contribute to reducing binding affinity and availability of taxanes. Second, taxane resistant tumors have increased stem cell like characteristics, indicating higher potential for further mutation in response to therapy. Third, the androgen receptor pathway is instrumental in the proliferation of CRPC and multiple hypotheses leading to this pathway reactivation have been reported. The connection of this pathway to the AKT pathway has received significant attention due to the upregulation of phosphorylated AKT in CRPC. This review highlights recent advances in elucidating taxane resistant mechanisms and summarizes potential therapeutic strategies for improved treatment of CRPC.
Collapse
|
41
|
Zhao HD, Xie HJ, Li J, Ren CP, Chen YX. Research Progress on Reversing Multidrug Resistance in Tumors by Using Chinese Medicine. Chin J Integr Med 2018; 24:474-480. [PMID: 29860581 DOI: 10.1007/s11655-018-2910-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 10/14/2022]
Abstract
Multidrug resistance (MDR) is a major cause of cancer chemotherapy failure, and it is important to develop suitable reversal agents to overcome MDR. A majority of chemical reversal agents have acceptable reversal effects. However, the toxicity and adverse reactions associated with these agents restricts their clinical use. Chinese medicines (CMs) have lower toxicities and adverse reactions and are associated with multiple components, multiple targets and reduced toxicity. CMs have several advantages and could reverse MDR, decrease drug dosage, enhance patient compliance and increase efficacy. This review summarizes the current progress of CM reversal agents..
Collapse
Affiliation(s)
- Huan-Dong Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, China.,School of Pharmacy, Central South University, Changsha, 410013, China
| | - Hong-Juan Xie
- Department of Pharmacy, Tongren Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200336, China
| | - Jian Li
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Cai-Ping Ren
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Yu-Xiang Chen
- School of Pharmacy, Central South University, Changsha, 410013, China.
| |
Collapse
|
42
|
Morikawa Y, Tagami T, Hoshikawa A, Ozeki T. The Use of an Efficient Microfluidic Mixing System for Generating Stabilized Polymeric Nanoparticles for Controlled Drug Release. Biol Pharm Bull 2018; 41:899-907. [DOI: 10.1248/bpb.b17-01036] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshinori Morikawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Akihiro Hoshikawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
43
|
Zhou H, Lv S, Zhang D, Deng M, Zhang X, Tang Z, Chen X. A polypeptide based podophyllotoxin conjugate for the treatment of multi drug resistant breast cancer with enhanced efficiency and minimal toxicity. Acta Biomater 2018; 73:388-399. [PMID: 29694920 DOI: 10.1016/j.actbio.2018.04.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
Abstract
Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown significant activity against P-glycoprotein (P-gp) mediated multi drug resistant cancer cells. However, because of the poor aqueous solubility and high toxicity, PPT cannot be used in clinical cancer therapy. In order to enhance the efficiency and reduce side effect of PPT, a polypeptide based PPT conjugate PLG-g-mPEG-PPT was developed and used for the treatment of multi drug resistant breast cancer. The PLG-g-mPEG-PPT was prepared by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol) (PLG-g-mPEG) via ester bonds. The PPT conjugates self-assembled into nanoparticles with average sizes about 100 nm in aqueous solution. Western blotting assay showed that the PLG-g-mPEG-PPT could effectively inhibit the expression of P-gp in the multiple drug resistant MCF-7/ADR cells. In vitro cytotoxicity assay indicated that the resistance index (RI) values of PLG-g-mPEG-PPT on different drug-resistant cancer cell lines exhibited 57-270 folds reduction than of traditional microtubule inhibitor chemotherapeutic drug PTX or DTX. Hemolysis assay demonstrated that the conjugation greatly decreased the hemolytic activity of free PPT. Maximum tolerated dose (MTD) of PLG-g-mPEG-PPT increased greatly (13.3 folds) as compared to that of free PPT. In vivo study showed that the PLG-g-mPEG-PPT conjugate remarkably enhanced the antitumor efficacy against MCF-7/ADR xenograft tumors with a tumor suppression rate (TSR) of 82.5%, displayed significantly improved anticancer efficacy as compared to free PPT (TSR = 37.1%) with minimal toxicity when both of the two formulations were used in MTD. STATEMENT OF SIGNIFICANCE The development of multiple drug resistance (MDR) of cancer cells is the main cause of chemotherapy failure. The over-expression of P-glycoprotein (P-gp) has been recognized to be the most important cause of MDR in cancer. Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown strong activity against P-gp mediated multidrug resistant cancer cells by simultaneously inhibiting the over-expression of P-gp and the growth of cancer cells. However, PPT can not be used in clinical cancer treatment due to its poor aqueous solubility and high toxicity. Herein, we developed a polypeptide based PPT conjugate PLG-g-mPEG-PPT by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol). The PLG-g-mPEG-PPT shows significantly decreased hemolytic activity, greatly improved maximum tolerated dose and remarkably enhanced antitumor efficacy against MCF-7/ADR xenograft tumors as compared to free PPT.
Collapse
|
44
|
Lan C, Zhao S. Self-assembled nanomaterials for synergistic antitumour therapy. J Mater Chem B 2018; 6:6685-6704. [DOI: 10.1039/c8tb01978a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recent progress on self-assembled nanodrugs for anticancer treatment was discussed.
Collapse
Affiliation(s)
- Chuanqing Lan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources
- Guangxi Normal University
- Guilin
- China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources
- Guangxi Normal University
- Guilin
- China
| |
Collapse
|
45
|
Cabazitaxel-conjugated nanoparticles for docetaxel-resistant and bone metastatic prostate cancer. Cancer Lett 2017; 410:169-179. [PMID: 28965854 DOI: 10.1016/j.canlet.2017.09.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Effective treatment of metastatic castration resistant prostate cancer (mCRPC) remains an unmet challenge. Cabazitaxel (CBZ) is approved for mCRPC after docetaxel (DTX) failure, but the improvement in survival is only moderate (∼2 months) and patients suffer from significant side effects. Here, we report the development of a polymer based delivery system for CBZ to improve its safety and efficacy against DTX-resistant mCRPC. CBZ was conjugated to a carboxymethylcellulose-based polymer (Cellax-CBZ), which self-assembled into ∼100 nm particles in saline and exhibited sustained drug release in serum at 10%/day. Cellax-CBZ delivered 157-fold higher CBZ to PC3-RES prostate tumor in mice and could be safely administered at a 25-fold higher dose compared to free CBZ, resulting in superior tumor inhibition in multiple mice models of DTX-resistant CRPC. In a metastatic bone model of CRPC, Cellax-CBZ significantly improves overall survival with a 70% long-term survival rate to day 120, while mice treated with free CBZ had a median survival of 40 days. Cellax-CBZ induced mild and reversible neutropenia in mice but no other tissue damage. Cellax-CBZ showed significant potential for improving therapy of mCRPC over clinically approved CBZ.
Collapse
|
46
|
Liu Y, Zhu S, Gu K, Guo Z, Huang X, Wang M, Amin HM, Zhu W, Shi P. GSH-Activated NIR Fluorescent Prodrug for Podophyllotoxin Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:29496-29504. [PMID: 28758393 DOI: 10.1021/acsami.7b07091] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Theranostic prodrug therapy enables the targeted delivery of anticancer drugs with minimized adverse effects and real-time in situ monitoring of activation of the prodrugs. In this work, we report the synthesis and biological assessment of the near-infrared (NIR) prodrug DCM-S-PPT and its amphiphilic copolymer (mPEG-DSPE)-encapsulated nanoparticles. DCM-S-PPT is composed of podophyllotoxin (PPT) as the anticancer moiety and a dicyanomethylene-4H-pyran (DCM) derivative as the NIR fluorescent reporter, which are linked by a thiol-specific cleavable disulfide bond. In vitro experiments indicated that DCM-S-PPT has low cytotoxicity and that glutathione (GSH) can activate DCM-S-PPT resulting in PPT release and a concomitant significant enhancement in NIR fluorescence at 665 nm. After being intravenously injected into tumor-bearing nude mice, DCM-S-PPT exhibited excellent tumor-activated performance. Furthermore, we have demonstrated that mPEG-DSPE as a nanocarrier loaded with DCM-S-PPT (mPEG-DSPE/DCM-S-PPT) showed even greater tumor-targeting performance than DCM-S-PPT on account of the enhanced permeability and retention effect. Its tumor-targeting ability and specific drug release in tumors make DCM-S-PPT a promising prodrug that could provide a significant strategy for theranostic drug delivery systems.
Collapse
Affiliation(s)
- Yajing Liu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| | - Shaojia Zhu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| | - Kaizhi Gu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| | - Zhiqian Guo
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| | - Xiaoyu Huang
- Key Laboratory of Synthetic and Self-Assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , 345 Lingling Road, Shanghai 200032, PR China
| | - Mingwei Wang
- Fudan University Shanghai Cancer Center , 270 Dongan Road, Shanghai 200032, PR China
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center , 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Weihong Zhu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Advanced Materials and Institute of Fine Chemicals, East China University of Science and Technology , 130 Meilong Road, Shanghai 200237, PR China
| |
Collapse
|
47
|
Roy A, Zhao Y, Yang Y, Szeitz A, Klassen T, Li SD. Selective targeting and therapy of metastatic and multidrug resistant tumors using a long circulating podophyllotoxin nanoparticle. Biomaterials 2017; 137:11-22. [PMID: 28528299 DOI: 10.1016/j.biomaterials.2017.05.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/20/2017] [Accepted: 05/09/2017] [Indexed: 01/01/2023]
Abstract
Treatment options for metastatic and multidrug resistant (MDR) tumors are limited, and most of the chemotherapeutic drugs exhibit low efficacy against MDR cancers. An anti-tubulin agent podophyllotoxin (PPT) displays high potency against MDR tumor cells. However, due to its poor solubility and non-specificity, PPT cannot be used systemically. We have developed a self-assembling nanoparticle dosage form for PPT (named Celludo) by covalently conjugating PPT and polyethylene glycol (PEG) to acetylated carboxymethyl cellulose (CMC-Ac) via ester linkages. Celludo displayed extended blood circulation with an 18-fold prolonged half-life (t1/2), 9000-fold higher area under the curve (AUC), and 1000-fold reduced clearance compared to free PPT. Tumor delivery was 500-fold higher in the Cellduo group compared to free PPT. Against the lung metastatic model of EMT6-AR1, Celludo showed selective localization in the metastatic nodules and increased the median survival to 20 d compared to 6-8 d with docetaxel and PPT treatment. In the intraperitoneal metastatic model of human ovarian NCI-ADR/RES tumor, Celludo prolonged the median survival from 50 d to 70 d, whereas the standard therapy PEGylated liposomal doxorubicin showed no effect. No major toxicity was detected with the Celludo treatment. These results demonstrate that Celludo is effective against metastatic and MDR tumors.
Collapse
Affiliation(s)
- Aniruddha Roy
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; Department of Pharmacy, Birla Institute of Technology & Science (BITS)-Pilani, Pilani Campus, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Yucheng Zhao
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Yang Yang
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Andras Szeitz
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Tara Klassen
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
48
|
Ling L, Yao C, Du Y, Ismail M, He R, Hou Y, Zhang Y, Li X. Assembled liposomes of dual podophyllotoxin phospholipid: preparation, characterization and in vivo anticancer activity. Nanomedicine (Lond) 2017; 12:657-672. [DOI: 10.2217/nnm-2016-0396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: A novel amphiphilic prodrug dual podophyllotoxin (PPT) succinate glycerophosphorylcholine (Di-PPT-GPC) assembled liposomes was developed to improve efficiency of PPT. Materials & methods: Di-PPT-GPC liposomes were prepared by thin film technique and characterized by dynamic light scattering and cryo-electron microscopy. Results: In vitro release studies showed that Di-PPT-GPC liposomes could significantly release PPT in weakly acidic environment but had good stability under biological conditions. Methyl tetrazolium assay data revealed that the liposomes have comparable cytotoxicities to free PPT against MCF-7, HeLa and U87 cells. More importantly, in vivo antitumor evaluation indicated that Di-PPT-GPC liposomes exhibited favorable tumor growth inhibition without side effects. Conclusion: Di-PPT-GPC liposomes might have potential to promote the therapeutic effect of PPT for cancer therapy.
Collapse
Affiliation(s)
- Longbing Ling
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chen Yao
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Muhammad Ismail
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ruiyu He
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yongpeng Hou
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ying Zhang
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| |
Collapse
|
49
|
Shen T, Xu X, Guo L, Tang H, Diao T, Gan Z, Zhang G, Yu Q. Efficient Tumor Accumulation, Penetration and Tumor Growth Inhibition Achieved by Polymer Therapeutics: The Effect of Polymer Architectures. Biomacromolecules 2016; 18:217-230. [DOI: 10.1021/acs.biomac.6b01533] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tong Shen
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Xin Xu
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Linyi Guo
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Hao Tang
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Tongxiang Diao
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Zhihua Gan
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| | - Guan Zhang
- Department
of Urology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Qingsong Yu
- The
State Key Laboratory of Organic−inorganic Composites, Beijing
Laboratory of Biomedical Materials, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People’s Republic of China
| |
Collapse
|
50
|
Yang Y, Bteich J, Li SD. Current Update of a Carboxymethylcellulose-PEG Conjugate Platform for Delivery of Insoluble Cytotoxic Agents to Tumors. AAPS JOURNAL 2016; 19:386-396. [PMID: 27873118 DOI: 10.1208/s12248-016-0014-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022]
Abstract
Cytotoxic chemotherapeutic agents are used as the standard therapy for a range of significant cancers, but many of these drugs suffer from poor water solubility and low selectivity, limiting their clinical efficacy. To overcome these shortcomings, Cellax™ drug delivery platform was developed. Cellax™ is a polymer-based nanoparticle drug delivery system designed to solubilize hydrophobic drugs and target them to solid tumors, thereby enhancing the efficacy and reducing the side effects. Cellax-docetaxel (Cellax-DTX) displayed improved pharmacokinetic, safety, and efficacy profiles compared to native DTX (Taxotere®) and Nab-paclitaxel (Nab-PTX, Abraxane®) in multiple animal models. Cellax-DTX was shown to interact with serum albumin and SPARC (secreted protein acidic and rich in cysteine) that is highly expressed by tumor stromal cells, leading to superior stroma depleting activity in orthotopic breast and pancreatic tumor models and subsequently reduced incidence of visceral metastases compared to free DTX and Nab-PTX. The Cellax™ platform was employed to deliver podophyllotoxin (Cellax-PPT) and cabazitaxel (Cellax-CBZ), and increased their safety and efficacy against multidrug-resistant tumors. This review discusses the rational design of the Cellax™ platform and summarizes the preclinical results. A multifunctional version of Cellax™ and a biomarker for predicting Cellax™ efficacy were developed and identified to promote the personalized use. Perspectives and future plans for this platform technology are also provided.
Collapse
Affiliation(s)
- Yang Yang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver Campus, 5519-2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Joseph Bteich
- Drug Delivery and Formulation, Drug Discovery Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario, M5G 0A3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver Campus, 5519-2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|