1
|
Cho Y, Lim J, Kim YH, Min J. ReV as a Novel S. cerevisiae-Derived Drug Carrier to Enhance Anticancer Therapy through Daunorubicin Delivery. Appl Biochem Biotechnol 2025; 197:2301-2311. [PMID: 39738861 DOI: 10.1007/s12010-024-05177-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 01/02/2025]
Abstract
This study explores the potential of vacuoles derived from Saccharomyces cerevisiae (S. cerevisiae) as a novel form of drug carrier, specifically focusing on their application in enhancing the delivery of the chemotherapeutic agent Daunorubicin (DNR). We isolated and reassembled these vacuoles, referred to as Reassembled Vacuoles (ReV), aiming to overcome the challenges of drug degradation caused by hydrolytic enzymes within traditional vacuoles. ReV encapsulating DNR were tested against HL-60 cells, a model for acute myeloid leukemia, to evaluate their therapeutic impact. Through various analyses, including Nanoparticle tracking analysis (NTA) and Field-emission electron scanning microscope (FE-SEM), we characterized the properties of ReV. Our findings revealed that ReV exhibited superior stability, drug release rate, and cytotoxic efficacy compared to normal vacuoles (NorV). Notably, ReV demonstrated a higher apoptosis rate in HL-60 cells, efficient and complete release of DNR within 24 h, and reduced cytotoxic side effects. These results suggest that ReV could represent a new and effective drug delivery system in anticancer therapy, paving the way for more targeted and safer cancer treatment modalities.
Collapse
Affiliation(s)
- Yunyoung Cho
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Jiwoo Lim
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University 1, Chungdae-Ro, Seowon-Gu, Cheongju, 28644, South Korea.
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea.
| |
Collapse
|
2
|
Saxena S, Sharma S, Kumar G, Thakur S. Unravelling the complexity of CARPA: a review of emerging advancements in therapeutic strategies. Arch Dermatol Res 2025; 317:439. [PMID: 39971823 DOI: 10.1007/s00403-025-03971-z] [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: 12/10/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
Hypersensitivity reactions to complement activation-related pseudo-allergy (CARPA) pose a serious concern to patient safety when using nanoparticle-based drug delivery systems (NDDS). Complement activation-related pseudo-allergy is a severe, idiosyncratic hypersensitivity reaction consequent to complement activation and liberation of potent pro-inflammatory molecules. Recent developments have concentrated on identifying, managing, and preventing CARPA to improve the efficacy and safety of NDDS, including early identification biomarkers and highly sensitive diagnostic techniques. The development of biocompatible nanoparticles, surface changes to reduce complement activation, and the use of complement inhibitors are some of the innovative strategies for CARPA reduction that are highlighted. Furthermore, newly developed management procedures, such as premedication schedules, customized dosage plans, and real-time monitoring methods, are also covered. The scope of this review encompasses the new diagnostic methods based on in-vitro assays, ex-vivo models, and highly sensitive imaging techniques for the detection of complement activation and other related pseudo-allergic reactions including liposome encapsulation and PEGylation to enhance biocompatibility and decrease immune stimulation. Special emphasis is paid to the application of high-throughput screening technologies and omics tools that enhance the likelihood and evaluation of CARPA immunogenicity. Integration of these approaches forms a comprehensive approach to improving the understanding and administration of CARPA in clinical settings to increase patient safety during nanoparticle-based treatment. The advanced alignments complement regulatory and clinical concerns and connect experimental paradigms to applications, such integration of knowledge provides a platform for the development of next-generation NDDSs for reducing CARPA and enhancing the efficiency of medication delivery thereby increasing patient compliance. This abstract delineates the methods of diagnosing, preventing, and managing CARPA, with addressing the nanotechnology for the problem.
Collapse
Affiliation(s)
- Shubhi Saxena
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Subhi Sharma
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Gourav Kumar
- Department of Pharmaceutics, ISF College of Pharmacy (An Autonomous College), Moga, Punjab, 142001, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy (An Autonomous College), Moga, Punjab, 142001, India.
| |
Collapse
|
3
|
Wu X, Wang F, Yang X, Gong Y, Niu T, Chu B, Qu Y, Qian Z. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403409. [PMID: 38934349 DOI: 10.1002/smll.202403409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Acute myeloid leukemia (AML) is a common and catastrophic hematological neoplasm with high mortality rates. Conventional therapies, including chemotherapy, hematopoietic stem cell transplantation (HSCT), immune therapy, and targeted agents, have unsatisfactory outcomes for AML patients due to drug toxicity, off-target effects, drug resistance, drug side effects, and AML relapse and refractoriness. These intrinsic limitations of current treatments have promoted the development and application of nanomedicine for more effective and safer leukemia therapy. In this review, the classification of nanoparticles applied in AML therapy, including liposomes, polymersomes, micelles, dendrimers, and inorganic nanoparticles, is reviewed. In addition, various strategies for enhancing therapeutic targetability in nanomedicine, including the use of conjugating ligands, biomimetic-nanotechnology, and bone marrow targeting, which indicates the potential to reverse drug resistance, are discussed. The application of nanomedicine for assisting immunotherapy is also involved. Finally, the advantages and possible challenges of nanomedicine for the transition from the preclinical phase to the clinical phase are discussed.
Collapse
Affiliation(s)
- Xia Wu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xijing Yang
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuping Gong
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
4
|
Shang Q, Chen Z, Li J, Guo M, Yang J, Jin Z, Shen Y, Guo S, Wang F. Two-pronged reversal of chemotherapy resistance by gold nanorods induced mild photothermal effect. Bioeng Transl Med 2024; 9:e10670. [PMID: 39553426 PMCID: PMC11561791 DOI: 10.1002/btm2.10670] [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: 09/03/2023] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 11/19/2024] Open
Abstract
Chemotherapy treatment outcomes are severely restricted by multidrug resistance (MDR), in which tumors develop a multiple cross-resistance toward drug involving the pump and nonpump resistance mechanisms, resulting in drug efflux and defending against drug toxicity. Herein, we constructed a pH and near infrared (NIR) light responsive nanomedicine DOX@FG based on gold nanorods (GNRs) that demonstrated the potential to improve chemotherapy outcomes by overcoming MDR. DOX@FG was constructed by conjugating folic acid (FA) and doxorubicin (DOX) derivatives onto GNRs, where the DOX derivatives possessed an acid-labile hydrazone bond. Stimulated by the acidic media in endocytic organelles, DOX@FG exhibited a responsive dissociation for the controlled release of chemotherapeutic DOX. Surprisingly, we found the mild photothermal effect elicited by GNRs under NIR irradiation simultaneously inhibited the pump and nonpump resistance mechanisms, enhancing the intracellular DOX accumulation and sensitizing the cancer cells to DOX, collectively amplify the chemotherapy efficacy and delay the MCF-7/ADR breast tumor growth. This intelligent DOX@FG nanomedicine with the potential for two-pronged reversal of MDR may provide a prospective way to encourage chemotherapy efficacy.
Collapse
Affiliation(s)
- Qi Shang
- School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Ziyan Chen
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jing Li
- Department of PharmacyPutuo People's HospitalShanghaiPeople's Republic of China
| | - Mingmei Guo
- School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jiapei Yang
- School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Zhu Jin
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Yuanyuan Shen
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| |
Collapse
|
5
|
Falanga AP, Massaro M, Borbone N, Notarbartolo M, Piccialli G, Liotta LF, Sanchez-Espejo R, Viseras Iborra C, Raymo FM, Oliviero G, Riela S. Carrier capability of halloysite nanotubes for the intracellular delivery of antisense PNA targeting mRNA of neuroglobin gene. J Colloid Interface Sci 2024; 663:9-20. [PMID: 38387188 DOI: 10.1016/j.jcis.2024.02.136] [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: 10/23/2023] [Revised: 12/28/2023] [Accepted: 02/17/2024] [Indexed: 02/24/2024]
Abstract
Peptide nucleic acid (PNA) is a DNA mimic that shows good stability against nucleases and proteases, forming strongly recognized complementary strands of DNA and RNA. However, due to its feeble ability to cross the cellular membrane, PNA activity and its targeting gene action is limited. Halloysite nanotubes (HNTs) are a natural and low-cost aluminosilicate clay. Because of their peculiar ability to cross cellular membrane, HNTs represent a valuable candidate for delivering genetic materials into cells. Herein, two differently charged 12-mer PNAs capable of recognizing as molecular target a 12-mer DNA molecule mimicking a purine-rich tract of neuroglobin were synthetized and loaded onto HNTs by electrostatic attraction interactions. After characterization, the kinetic release was also assessed in media mimicking physiological conditions. Resonance light scattering measurements assessed their ability to bind complementary single-stranded DNA. Furthermore, their intracellular delivery was assessed by confocal laser scanning microscopy on living MCF-7 cells incubated with fluorescence isothiocyanate (FITC)-PNA and HNTs labeled with a probe. The nanomaterials were found to cross cellular membrane and cell nuclei efficiently. Finally, it is worth mentioning that the HNTs/PNA can reduce the level of neuroglobin gene expression, as shown by reverse transcription-quantitative polymerase chain reaction and western blotting analysis.
Collapse
Affiliation(s)
- Andrea P Falanga
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Marina Massaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128 Palermo, Italy
| | - Nicola Borbone
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | - Monica Notarbartolo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, 90128 Palermo, Italy
| | - Gennaro Piccialli
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Leonarda F Liotta
- Istituto per lo Studio dei Materiali Nanostrutturati (ISMN)-CNR, Via Ugo La Malfa 153, Palermo 90146, Italy
| | - Rita Sanchez-Espejo
- University of Granada, Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, 18071 Granada, Spain
| | - Cesar Viseras Iborra
- University of Granada, Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, 18071 Granada, Spain; Andalusian Institute of Earth Sciences, CSIC-UGR, 18100 Armilla, Granada, Spain
| | - Françisco M Raymo
- Laboratory for Molecular Photonics, Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables 33146-0431, FL, United States
| | - Giorgia Oliviero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Serena Riela
- Dipartimento di Scienze Chimiche, Viale Andrea Doria 6, 95125 Catania, Italy.
| |
Collapse
|
6
|
Cavanagh RJ, Monteiro PF, Moloney C, Travanut A, Mehradnia F, Taresco V, Rahman R, Martin SG, Grabowska AM, Ashford MB, Alexander C. Free drug and ROS-responsive nanoparticle delivery of synergistic doxorubicin and olaparib combinations to triple negative breast cancer models. Biomater Sci 2024; 12:1822-1840. [PMID: 38407276 DOI: 10.1039/d3bm01931d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Combinations of the topoisomerase II inhibitor doxorubicin and the poly (ADP-ribose) polymerase inhibitor olaparib offer potential drug-drug synergy for the treatment of triple negative breast cancers (TNBC). In this study we performed in vitro screening of combinations of these drugs, administered directly or encapsulated within polymer nanoparticles, in both 2D and in 3D spheroid models of breast cancer. A variety of assays were used to evaluate drug potency, and calculations of combination index (CI) values indicated that synergistic effects of drug combinations occurred in a molar-ratio dependent manner. It is suggested that the mechanisms of synergy were related to enhancement of DNA damage as shown by the level of double-strand DNA breaks, and mechanisms of antagonism associated with mitochondrial mediated cell survival, as indicated by reactive oxygen species (ROS) generation. Enhanced drug delivery and potency was observed with nanoparticle formulations, with a greater extent of doxorubicin localised to cell nuclei as evidenced by microscopy, and higher cytotoxicity at the same time points compared to free drugs. Together, the work presented identifies specific combinations of doxorubicin and olaparib which were most effective in a panel of TNBC cell lines, explores the mechanisms by which these combined agents might act, and shows that formulation of these drug combinations into polymeric nanoparticles at specific ratios conserves synergistic action and enhanced potency in vitro compared to the free drugs.
Collapse
Affiliation(s)
| | - Patrícia F Monteiro
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Cara Moloney
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | | | | | | | - Ruman Rahman
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Stewart G Martin
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Anna M Grabowska
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Marianne B Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | | |
Collapse
|
7
|
Yang M, Chen Y, Zhu L, You L, Tong H, Meng H, Sheng J, Jin J. Harnessing Nanotechnology: Emerging Strategies for Multiple Myeloma Therapy. Biomolecules 2024; 14:83. [PMID: 38254683 PMCID: PMC10813273 DOI: 10.3390/biom14010083] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Advances in nanotechnology have provided novel avenues for the diagnosis and treatment of multiple myeloma (MM), a hematological malignancy characterized by the clonal proliferation of plasma cells in the bone marrow. This review elucidates the potential of nanotechnology to revolutionize myeloma therapy, focusing on nanoparticle-based drug delivery systems, nanoscale imaging techniques, and nano-immunotherapy. Nanoparticle-based drug delivery systems offer enhanced drug targeting, reduced systemic toxicity, and improved therapeutic efficacy. We discuss the latest developments in nanocarriers, such as liposomes, polymeric nanoparticles, and inorganic nanoparticles, used for the delivery of chemotherapeutic agents, siRNA, and miRNA in MM treatment. We delve into nanoscale imaging techniques which provide spatial multi-omic data, offering a holistic view of the tumor microenvironment. This spatial resolution can help decipher the complex interplay between cancer cells and their surrounding environment, facilitating the development of highly targeted therapies. Lastly, we explore the burgeoning field of nano-immunotherapy, which employs nanoparticles to modulate the immune system for myeloma treatment. Specifically, we consider how nanoparticles can be used to deliver tumor antigens to antigen-presenting cells, thus enhancing the body's immune response against myeloma cells. In conclusion, nanotechnology holds great promise for improving the prognosis and quality of life of MM patients. However, several challenges remain, including the need for further preclinical and clinical trials to assess the safety and efficacy of these emerging strategies. Future research should also focus on developing personalized nanomedicine approaches, which could tailor treatments to individual patients based on their genetic and molecular profiles.
Collapse
Affiliation(s)
- Min Yang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Yu Chen
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Li Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Liangshun You
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Haitao Meng
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Jianpeng Sheng
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (M.Y.); (Y.C.); (L.Z.); (L.Y.); (H.T.); (H.M.)
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou 310027, China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310029, China;
| |
Collapse
|
8
|
Stepanova M, Nikiforov A, Tennikova T, Korzhikova-Vlakh E. Polypeptide-Based Systems: From Synthesis to Application in Drug Delivery. Pharmaceutics 2023; 15:2641. [PMID: 38004619 PMCID: PMC10674432 DOI: 10.3390/pharmaceutics15112641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Synthetic polypeptides are biocompatible and biodegradable macromolecules whose composition and architecture can vary over a wide range. Their unique ability to form secondary structures, as well as different pathways of modification and biofunctionalization due to the diversity of amino acids, provide variation in the physicochemical and biological properties of polypeptide-containing materials. In this review article, we summarize the advances in the synthesis of polypeptides and their copolymers and the application of these systems for drug delivery in the form of (nano)particles or hydrogels. The issues, such as the diversity of polypeptide-containing (nano)particle types, the methods for their preparation and drug loading, as well as the influence of physicochemical characteristics on stability, degradability, cellular uptake, cytotoxicity, hemolysis, and immunogenicity of polypeptide-containing nanoparticles and their drug formulations, are comprehensively discussed. Finally, recent advances in the development of certain drug nanoformulations for peptides, proteins, gene delivery, cancer therapy, and antimicrobial and anti-inflammatory systems are summarized.
Collapse
Affiliation(s)
- Mariia Stepanova
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| | - Alexey Nikiforov
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| | - Tatiana Tennikova
- Institute of Chemistry, Saint-Petersburg State University, Universitetskiy pr. 26, Petergof, 198504 St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| |
Collapse
|
9
|
Zashikhina N, Gandalipov E, Dzhuzha A, Korzhikov-Vlakh V, Korzhikova-Vlakh E. Dual drug loaded polypeptide delivery systems for cancer therapy. J Microencapsul 2023:1-19. [PMID: 37824702 DOI: 10.1080/02652048.2023.2270064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The present study was aimed to prepare and examine in vitro novel dual-drug loaded delivery systems. Biodegradable nanoparticles based on poly(L-glutamic acid-co-D-phenylalanine) were used as nanocarriers for encapsulation of two drugs from the paclitaxel, irinotecan, and doxorubicin series. The developed delivery systems were characterised with hydrodynamic diameters less than 300 nm (PDI < 0.3). High encapsulation efficiencies (≥75%) were achieved for all single- and dual-drug formulations. The release studies showed faster release at acidic pH, with the release rate decreasing over time. The release patterns of the co-encapsulated forms of substances differed from those of the separately encapsulated drugs, suggesting differences in drug-polymer interactions. The joint action of encapsulated drugs was analysed using the colon cancer cells, both for the dual-drug delivery sytems and a mixture of single-drug formulations. The encapsulated forms of the drug combinations demonstrated comparable efficacy to the free forms, with the encapsulation enhancing solubility of the hydrophobic drug paclitaxel.
Collapse
Affiliation(s)
- Natalia Zashikhina
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
| | - Erik Gandalipov
- International Institute of Solution Chemistry and Advanced Materials Technologies, ITMO University, St. Petersburg, Russia
| | - Apollinariia Dzhuzha
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| | - Viktor Korzhikov-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Chemistry, Saint-Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
10
|
Rajeshkumar RR, Pavadai P, Panneerselvam T, Deepak V, Pandian SRK, Kabilan SJ, Vellaichamy S, Jeyaraman A, Kumar ASK, Sundar K, Kunjiappan S. Glucose-conjugated glutenin nanoparticles for selective targeting and delivery of camptothecin into breast cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2571-2586. [PMID: 37022437 DOI: 10.1007/s00210-023-02480-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
Receptor-mediated drug delivery systems are a promising tool for targeting malignant cells to suppress/inhibit the malignancy without disturbing healthy cells. Protein-based nanocarrier systems possess numerous advantages for the delivery of variety of chemotherapeutics, including therapeutic peptides and genes. In the present work, glucose-conjugated camptothecin-loaded glutenin nanoparticles (Glu-CPT-glutenin NPs) were fabricated to deliver camptothecin to MCF-7 cells via GLUT-1 transporter protein. Initially, Glu-conjugated glutenin polymer was successfully synthesized through reductive amination reaction, and this was confirmed by FTIR and 13C-NMR. Then, camptothecin (CPT) was loaded into Glu-conjugated glutenin polymer forming Glu-CPT-glutenin NPs. The nanoparticles were studied for their drug releasing capacity, morphological shape, size, physical nature, and zeta potential. The fabricated Glu-CPT-glutenin NPs were found to be spherical in shape and amorphous in nature with 200-nm size range and a zeta potential of - 30 mV. Furthermore, MTT assay using Glu-CPT-glutenin NPs confirmed concentration-dependent cytotoxicity against MCF-7 cells after 24-h treatment, and IC50 was found to be 18.23 μg mL-1. In vitro cellular uptake study demonstrated that the Glu-CPT-glutenin NPs had enhanced endocytosis and delivered CPT in MCF-7 cells. A typical apoptotic morphological change of condensed nuclei and distorted membrane bodies was found after treatment with IC50 concentration of NPs. The released CPT from NPs also targeted mitochondria of MCF-7 cells, significantly increasing the level of reactive oxygen species and causing the damage of mitochondrial membrane integrity. These outcomes confirmed that the wheat glutenin can positively serve as a significant delivery vehicle and enhance the anticancer potential of this drug.
Collapse
Affiliation(s)
- Raja Rajeswari Rajeshkumar
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Virudhunagar, 626126, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru, 560054, India
| | - Theivendren Panneerselvam
- Department of Pharmaceutical Chemistry, Swamy Vivekanandha College of Pharmacy, Elayampalayam, Namakkal, 637205, India
| | - Venkataraman Deepak
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Virudhunagar, 626126, India
- Maternal and Fetal Health Research Centre, 5Th Floor St. Mary's Hospital, University of Manchester, Oxford Road, Manchester, M13 9WL, UK
| | - Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Virudhunagar, 626126, India
| | | | - Sivakumar Vellaichamy
- Department of Pharmaceutics, Arulmigu Kalasalingam College of Pharmacy, Krishnankoil, Virudhunagar, 626126, India
| | - Anbu Jeyaraman
- Department of Pharmacology, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru, 560054, India
| | - A Santhana Krishna Kumar
- Department of Chemistry, National Sun Yat-Sen University, Gushan District, No. 70, Lien-Hai Road, Kaohsiung, 80424, Taiwan
- Faculty of Geology, Geophysics and Environmental Protection, AGH University of Science and Technology, 30-059, Krakow, Poland
| | - Krishnan Sundar
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Virudhunagar, 626126, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, Virudhunagar, 626126, India.
| |
Collapse
|
11
|
Lv L, Zhou X. Targeting Hippo signaling in cancer: novel perspectives and therapeutic potential. MedComm (Beijing) 2023; 4:e375. [PMID: 37799806 PMCID: PMC10547939 DOI: 10.1002/mco2.375] [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: 04/19/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
As highly conserved among diverse species, Hippo signaling pathway regulates various biological processes, including development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size. Studies in the last two decades have provided a good framework for how these fundamental functions of Hippo signaling are tightly regulated by a network with numerous intracellular and extracellular factors. The Hippo signaling pathway, when dysregulated, may lead to a wide variety of diseases, especially cancer. There is growing evidence demonstrating that dysregulated Hippo signaling is closely associated with tumorigenesis, cancer cell invasion, and migration, as well as drug resistance. Therefore, the Hippo pathway is considered an appealing therapeutic target for the treatment of cancer. Promising novel agents targeting the Hippo signaling pathway for cancers have recently emerged. These novel agents have shown antitumor activity in multiple cancer models and demonstrated therapeutic potential for cancer treatment. However, the detailed molecular basis of the Hippo signaling-driven tumor biology remains undefined. Our review summarizes current advances in understanding the mechanisms by which Hippo signaling drives tumorigenesis and confers drug resistance. We also propose strategies for future preclinical and clinical development to target this pathway.
Collapse
Affiliation(s)
- Liemei Lv
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
12
|
Dabke A, Ghosh S, Dabke P, Sawant K, Khopade A. Revisiting the in-vitro and in-vivo considerations for in-silico modelling of complex injectable drug products. J Control Release 2023; 360:185-211. [PMID: 37353161 DOI: 10.1016/j.jconrel.2023.06.029] [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: 01/28/2023] [Revised: 05/24/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Complex injectable drug products (CIDPs) have often been developed to modulate the pharmacokinetics along with efficacy for therapeutic agents used for remediation of chronic disorders. The effective development of CIDPs has exhibited complex kinetics associated with multiphasic drug release from the prepared formulations. Consequently, predictability of pharmacokinetic modelling for such CIDPs has been difficult and there is need for advanced complex computational models for the establishment of accurate prediction models for in-vitro-in-vivo correlation (IVIVC). The computational modelling aims at supplementing the existing knowledge with mathematical equations to develop formulation strategies for generation of predictable and discriminatory IVIVC. Such an approach would help in reduction of the burden of effect of hidden factors on preclinical to clinical translations. Computational tools like physiologically based pharmacokinetics (PBPK) modelling have combined physicochemical and physiological properties along with IVIVC characteristics of clinically used formulations. Such techniques have helped in prediction and understanding of variability in pharmacodynamic parameters of potential generic products to clinically used formulations like Doxil®, Ambisome®, Abraxane® in healthy and diseased population using mathematical equations. The current review highlights the important formulation characteristics, in-vitro, preclinical in-vivo aspects which need to be considered while developing a stimulatory predictive PBPK model in establishment of an IVIVC and in-vitro-in-vivo relationship (IVIVR).
Collapse
Affiliation(s)
- Amit Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Biopharmaceutics, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India
| | - Saikat Ghosh
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Pallavi Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Krutika Sawant
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India.
| | - Ajay Khopade
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India.
| |
Collapse
|
13
|
Liang X, Yang Y, Huang C, Ye Z, Lai W, Luo J, Li X, Yi X, Fan JB, Wang Y, Wang Y. cRGD-targeted heparin nanoparticles for effective dual drug treatment of cisplatin-resistant ovarian cancer. J Control Release 2023; 356:691-701. [PMID: 36933699 DOI: 10.1016/j.jconrel.2023.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 03/20/2023]
Abstract
Resistance to the chemotherapeutic agent cisplatin (DDP) is the primary reason for invalid chemotherapy of ovarian cancer. Given the complex mechanisms underlying chemo-resistance, the design of combination therapies based on blocking multiple mechanisms is a rationale to synergistically elevate therapeutic effect for effectively overcoming cancer chemo-resistance. Herein, we demonstrated a multifunctional nanoparticle (DDP-Ola@HR), which could simultaneously co-deliver DDP and Olaparib (Ola, DNA damage repair inhibitor) using targeted ligand cRGD peptide modified with heparin (HR) as nanocarrier, enabling the concurrent tackling of multiple resistance mechanisms to effectively inhibit the growth and metastasis of DDP-resistant ovarian cancer. In combination strategy, heparin could suppress the function of multidrug resistance-associated protein 2 (MRP2) and P-glycoprotein (P-gp) to promote the intracellular accumulation of DDP and Ola by specifically binding with heparanase (HPSE) to down-regulate PI3K/AKT/mTOR signaling pathway, and simultaneously served as a carrier combined with Ola to synergistically enhance the anti-proliferation ability of DDP for resistant ovarian cancer, thus achieving great therapeutic efficacy. Our DDP-Ola@HR could provide a simple and multifunctional combination strategy to trigger an anticipated cascading effect, thus effectively overcoming the chemo-resistance of ovarian cancer.
Collapse
Affiliation(s)
- Xiaomei Liang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Yulu Yang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Chuanqing Huang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Zhibin Ye
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Wujiang Lai
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jiamao Luo
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiaoxuan Li
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiao Yi
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jun-Bing Fan
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Ying Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Yifeng Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| |
Collapse
|
14
|
Muthwill MS, Kong P, Dinu IA, Necula D, John C, Palivan CG. Tailoring Polymer-Based Nanoassemblies for Stimuli-Responsive Theranostic Applications. Macromol Biosci 2022; 22:e2200270. [PMID: 36100461 DOI: 10.1002/mabi.202200270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/28/2022] [Indexed: 12/25/2022]
Abstract
Polymer assemblies on the nanoscale represent a powerful toolbox for the design of theranostic systems when combined with both therapeutic compounds and diagnostic reporting ones. Here, recent advances in the design of theranostic systems for various diseases, containing-in their architecture-either polymers or polymer assemblies as one of the building blocks are presented. This review encompasses the general principles of polymer self-assembly, from the production of adequate copolymers up to supramolecular assemblies with theranostic functionality. Such polymer nanoassemblies can be further tailored through the incorporation of inorganic nanoparticles to endow them with multifunctional therapeutic and/or diagnostic features. Systems that change their architecture or properties in the presence of stimuli are selected, as responsivity to changes in the environment is a key factor for enhancing efficiency. Such theranostic systems are based on the intrinsic properties of copolymers or one of the other components. In addition, systems with a more complex architecture, such as multicompartments, are presented. Selected systems indicate the advantages of such theranostic approaches and provide a basis for further developments in the field.
Collapse
Affiliation(s)
- Moritz S Muthwill
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland.,NCCR-Molecular Systems Engineering, Mattenstrasse 24a, BPR 1095, Basel, 4058, Switzerland
| | - Phally Kong
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland
| | - Ionel Adrian Dinu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland
| | - Danut Necula
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland
| | - Christoph John
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland.,NCCR-Molecular Systems Engineering, Mattenstrasse 24a, BPR 1095, Basel, 4058, Switzerland
| |
Collapse
|
15
|
Ding Y, Wang C, Ma Y, Zhu L, Lu B, Wang Y, Wang J, Dong CM, Yao Y. Tumor microenvironment responsive polypeptide-based supramolecular nanoprodrugs for combination therapy. Acta Biomater 2022. [DOI: 10.10.1016/j.actbio.2022.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Ding Y, Wang C, Ma Y, Zhu L, Lu B, Wang Y, Wang J, Dong CM, Yao Y. Tumor microenvironment responsive polypeptide-based supramolecular nanoprodrugs for combination therapy. Acta Biomater 2022; 146:396-405. [PMID: 35470074 DOI: 10.1016/j.actbio.2022.04.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022]
Abstract
Tumor microenvironment responsive nanomedicine has drawn considerable attention for combination therapy, but still remains a significant challenge for less side effects and enhanced anti-tumor efficiency. Herein, we develop a pH/ROS dual-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) by using pillar[5]arene-based host-guest strategy for combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT). The PFW-DOX/GOD consists of a pH-responsive ferrocene/pillar[5]arene-containing polypeptide, a ROS-responsive polyprodrug, and encapsulated glucose oxidase (GOD). Upon into intracellular acidic environment, PFW-DOX/GOD exhibits rapid pH-triggered disassembly behavior. Simultaneously, the released GOD can catalyze intratumoral glucose into massive H2O2, which are further converted into highly toxic hydroxyl radicals (•OH) by the catalysis of ferrocene via the Fenton reaction. Thereafter, induced by the ROS-responsive cleavage of thioketal linkage, the conjugated DOX prodrug was released and activated. The combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT) of PFW-DOX/GOD present anti-tumor effect with 96% of tumor inhibitory rate (TIR). Therefore, such tumor microenvironment-responsive supramolecular polypeptide nanoprodrugs represent a potential candidate for combination therapy with minimal side effects. STATEMENT OF SIGNIFICANCE: In this work, a tumor microenvironment-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) was prepared via pillar[5]arene-based host-guest interactions, and presented low side effects and high tumor accumulation owing to the diameters of about 200 nm and surface PEG segment. After pH-responsive release of GOD in the intracellular acidic environment, the cascade catalytic reactions including GOD-catalyzed degradation of intratumoral glucose and Fenton reaction, effectively happened to generate •OH for chemodynamic therapy (CDT), which subsequently induced the cleavage of thioketal linkage to activate free DOX for chemotherapy (CT). Collectively, this supramolecular polypeptide nanoprodrugs provide a promising strategy for combination therapy with synergetic anti-tumor effect.
Collapse
Affiliation(s)
- Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China.
| | - Chenwei Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Yuxuan Ma
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Lvming Zhu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Bing Lu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Jin Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Chang-Ming Dong
- Joint Research Center for Precision Medicine, School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China.
| |
Collapse
|
17
|
Javia A, Vanza J, Bardoliwala D, Ghosh S, Misra A, Patel M, Thakkar H. Polymer-drug conjugates: Design principles, emerging synthetic strategies and clinical overview. Int J Pharm 2022; 623:121863. [PMID: 35643347 DOI: 10.1016/j.ijpharm.2022.121863] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/06/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
Adagen, an enzyme replacement treatment for adenosine deaminase deficiency, was the first protein-polymer conjugate to be approved in early 1990s. Post this regulatory approval, numerous polymeric drugs and polymeric nanoparticles have entered the market as advanced or next-generation polymer-based therapeutics, while many others have currently been tested clinically. The polymer conjugation to therapeutic moiety offers several advantages, like enhanced solubilization of drug, controlled release, reduced immunogenicity, and prolonged circulation. The present review intends to highlight considerations in the design of therapeutically effective polymer-drug conjugates (PDCs), including the choice of linker chemistry. The potential synthetic strategies to formulate PDCs have been discussed along with recent advancements in the different types of PDCs, i.e., polymer-small molecular weight drug conjugates, polymer-protein conjugates, and stimuli-responsive PDCs, which are under clinical/preclinical investigation. Current impediments and regulatory hurdles hindering the clinical translation of PDC into effective therapeutic regimens for the amelioration of disease conditions have been addressed.
Collapse
Affiliation(s)
- Ankit Javia
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Jigar Vanza
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat-388421, India
| | - Denish Bardoliwala
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India; Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKM's NMIMS, Shirpur, Maharashtra-425405, Indi
| | - Mrunali Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat-388421, India
| | - Hetal Thakkar
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India.
| |
Collapse
|
18
|
Mahajan S, Aalhate M, Guru SK, Singh PK. Nanomedicine as a magic bullet for combating lymphoma. J Control Release 2022; 347:211-236. [PMID: 35533946 DOI: 10.1016/j.jconrel.2022.05.002] [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] [Received: 02/23/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Hematological malignancy like lymphoma originates in lymph tissues and has a propensity to spread across other organs. Managing such tumors is challenging as conventional strategies like surgery and local treatment are not plausible options and there are high chances of relapse. The advent of novel targeted therapies and antibody-mediated treatments has proven revolutionary in the management of these tumors. Although these therapies have an added advantage of specificity in comparison to the traditional chemotherapy approach, such treatment alternatives suffer from the occurrence of drug resistance and dose-related toxicities. In past decades, nanomedicine has emerged as an excellent surrogate to increase the bioavailability of therapeutic moieties along with a reduction in toxicities of highly cytotoxic drugs. Nanotherapeutics achieve targeted delivery of the therapeutic agents into the malignant cells and also have the ability to carry genes and therapeutic proteins to the desired sites. Furthermore, nanomedicine has an edge in rendering personalized medicine as one type of lymphoma is pathologically different from others. In this review, we have highlighted various applications of nanotechnology-based delivery systems based on lipidic, polymeric and inorganic nanomaterials that address different targets for effectively tackling lymphomas. Moreover, we have discussed recent advances and therapies available exclusively for managing this malignancy.
Collapse
Affiliation(s)
- Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
19
|
Ghosh S, Mishra P, Banerjee S, Maiti K, Khopade A, Misra A, Sawant K, Bhowmick S. Exploration of the cardinal formulation parameters influencing the encapsulation and physicochemical properties of co-loaded anticancer dual drug nanoliposomes. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Xiao J, Lu Y, Lu D, Chen W, Hu W, Zhao Y, Chen S. Co‐delivery of paclitaxel and
CXCL1 shRNA
via cationic polymeric micelles for synergistic therapy against ovarian cancer. POLYM INT 2022. [DOI: 10.1002/pi.6406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jingjing Xiao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yingying Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Deng Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Wulian Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai 200433 PR China
| | - Weiguo Hu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yuqing Zhao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Shouzhen Chen
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| |
Collapse
|
21
|
Ci T, Zhang W, Qiao Y, Li H, Zang J, Li H, Feng N, Gu Z. Delivery strategies in treatments of leukemia. Chem Soc Rev 2022; 51:2121-2144. [PMID: 35188506 DOI: 10.1039/d1cs00755f] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukemia is a hematological malignancy associated with the uncontrolled proliferation of mutant progenitors, suppressing the production of normal blood cells. Current treatments, including chemotherapy, radiotherapy, and immunotherapy, still lead to unsatisfactory results with a 5 year survival rate of only 30-50%. The poor prognosis is related to both disease relapse and treatment-associated toxicity. Delivery strategies can improve the in vivo pharmacokinetics of drugs, navigating the therapeutics to target cells or the tumor microenvironment and reversing drug resistance, which maximizes tumor elimination and alleviates systematic adverse effects. This review discusses available FDA-approved anti-leukemia drugs and therapies with a focus on the advances in the development of anti-leukemia drug delivery systems. Additionally, challenges in clinical translation of the delivery strategies and future research opportunities in leukemia treatment are also included.
Collapse
Affiliation(s)
- Tianyuan Ci
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wentao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingyu Qiao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jing Zang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
22
|
Au-O-MWCNTs and TiO2-O-MWCNTs as Efficient Nanocarriers for Dexamethasone: Adsorption Isotherms and Kinetic Studies. INTERNATIONAL JOURNAL OF CHEMICAL ENGINEERING 2021. [DOI: 10.1155/2021/2040363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this research, the fabrication of drug delivery systems based on oxidized multiwall carbon nanotubes (O-MWCNTs) was studied. Herein, TiO2 and Au were conjugated with O-MWCNTs to prepare efficient nanocarriers for dexamethasone (dex). The samples were characterized by Fourier transform infrared (FTIR), scanning electron microscopy (SEM), and X-ray diffraction (XRD). In addition, dex loading was studied using adsorption isotherms including Langmuir, Freundlich, Temkin, and Dubinin–Radushkevich. The results show that dex adsorption agreed well with the Freundlich isotherm. Increasing the TiO2 to O-MWCNT ratio from (1 : 4) to (1 : 2) can improve the adsorption capacity from
to 320
. The increasing Au amount increases the adsorption capacity from
(SA1) to maximum
(SA6). The maximum equilibrium binding energy
was obtained for SA2, and SA7 shows high binding strength between dex and the nanoadsorbent. Carbon nanotubes (CNTs) show good affinity with high loading capabilities for dexamethasone adsorption. The synthesized TiO2-O-MWCNTs:1/2 with the maximum removal percent (80%) was proposed as an appropriate nanocarrier for dexamethasone. Pseudo-first order, pseudo-second order, Elovich, and intraparticle diffusion models were investigated for all synthesized drug nanocarriers. According to regression coefficients, experimental data are in good agreement with the pseudo-second order model for all adsorbents except O-MWCNT/CTAB. Experimental results revealed that the Elovich model could account for the O-MWCNT/CTAB adsorbent.
Collapse
|
23
|
Su Z, Dong S, Zhao SC, Liu K, Tan Y, Jiang X, Assaraf YG, Qin B, Chen ZS, Zou C. Novel nanomedicines to overcome cancer multidrug resistance. Drug Resist Updat 2021; 58:100777. [PMID: 34481195 DOI: 10.1016/j.drup.2021.100777] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022]
Abstract
Chemotherapy remains a powerful tool to eliminate malignant cells. However, the efficacy of chemotherapy is compromised by the frequent emergence of intrinsic and acquired multidrug resistance (MDR). These chemoresistance modalities are based on a multiplicity of molecular mechanisms of drug resistance, including : 1) Impaired drug uptake into cancer cells; 2) Increased expression of ATP-binding cassette efflux transporters; 3) Loss of function of pro-apoptotic factors; 4) Enhanced DNA repair capacity; 5) Qualitative or quantitative alterations of specific cellular targets; 6) Alterations that allow cancer cells to tolerate adverse or stressful conditions; 7) Increased biotransformation or metabolism of anticancer drugs to less active or completely inactive metabolites; and 8) Intracellular and intercellular drug sequestration in well-defined organelles away from the cellular target. Hence, one of the major aims of cancer research is to develop novel strategies to overcome cancer drug resistance. Over the last decades, nanomedicine, which focuses on targeted delivery of therapeutic drugs into tumor tissues using nano-sized formulations, has emerged as a promising tool for cancer treatment. Therefore, nanomedicine has been introduced as a reliable approach to improve treatment efficacy and minimize detrimental adverse effects as well as overcome cancer drug resistance. With rationally designed strategies including passively targeted delivery, actively targeted delivery, delivery of multidrug combinations, as well as multimodal combination therapy, nanomedicine paves the way towards efficacious cancer treatment and hold great promise in overcoming cancer drug resistance. Herein, we review the recent progress of nanomaterials used in medicine, including liposomal nanoparticles, polymeric nanoparticles, inorganic nanoparticles and hybrid nanoparticles, to surmount cancer multidrug resistance. Finally, the future perspectives of the application of nanomedicine to reverse cancer drug resistance will be addressed.
Collapse
Affiliation(s)
- Zhenwei Su
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, PR China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, 518001, Guangdong, PR China
| | - Shaowei Dong
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, PR China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, 518001, Guangdong, PR China
| | - Shan-Chao Zhao
- Department of Urology, the Third Affiliated Hospital of Southern Medical University; Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, PR China
| | - Kaisheng Liu
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, PR China
| | - Yao Tan
- Shenzhen Aier Eye Hospital, Jinan University, No. 2048, Huaqiang South Road, Futian District, Shenzhen, 518032, Guangdong, PR China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Bo Qin
- Shenzhen Aier Eye Hospital, Jinan University, No. 2048, Huaqiang South Road, Futian District, Shenzhen, 518032, Guangdong, PR China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, 11439, New York, USA.
| | - Chang Zou
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, PR China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, 518001, Guangdong, PR China.
| |
Collapse
|
24
|
Triple negative breast cancer and non-small cell lung cancer: Clinical challenges and nano-formulation approaches. J Control Release 2021; 337:27-58. [PMID: 34273417 DOI: 10.1016/j.jconrel.2021.07.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) are amongst the most aggressive forms of solid tumors. TNBC is highlighted by absence of genetic components of progesterone receptor, HER2/neu and estrogen receptor in breast cancer. NSCLC is characterized by integration of malignant carcinoma into respiratory system. Both cancers are associated with poor median and overall survival rates with low progression free survival with high incidences of relapse. These cancers are characterized by tumor heterogeneity, genetic mutations, generation of cancer-stem cells, immune-resistance and chemoresistance. Further, these neoplasms have been reported for tumor cross-talk into second primary cancers for each other. Current chemotherapeutic regimens include usage of multiple agents in tandem to affect tumor cells through multiple mechanisms with various such combinations being clinically tested. However, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Consequently, passive targeted albumin bound paclitaxel and PEGylated liposomal doxorubicin have been clinically used and tested with newer drugs for improved therapeutic efficacy in these cancers. Active targeting of nanocarriers against surface overexpressed proteins in both neoplasms have been explored. However, use of single agent nanoparticulate formulations against both cancers have failed to elicit desired outcomes. This review aims to identify clinical unmet need in these cancers while establishing a correlation with tested nano-formulation approaches and issues with preclinical to clinical translation. Lipid and polymer-based drug-drug and drug-gene combinatorial nanocarriers delivering multiple chemotherapeutics simultaneously to desired site of action have been detailed. Finally, emerging opportunities such as pharmacological targets (immune check point and epigentic modulators) as well as gene-based modulation (siRNA/CRISPR/Cas9) and the nano-formulation challenges for effective treatment of both cancers have been explored.
Collapse
|
25
|
Formulation and clinical perspectives of inhalation-based nanocarrier delivery: a new archetype in lung cancer treatment. Ther Deliv 2021; 12:397-418. [PMID: 33902294 DOI: 10.4155/tde-2020-0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite tremendous research in targeted delivery and specific molecular inhibitors (gene delivery), cytotoxic drug delivery through inhalation has been seen as a core part in the treatment of the lung cancer. Inhalation delivery provides a high dose of the drug directly to the lungs without affecting other body organs, increasing the therapeutic ratio. This article reviews the research performed over the last several decades regarding inhalation delivery of various cancer therapeutics for the treatment of lung cancer. Nevertheless, pulmonary administration of nanocarrier-based cancer therapeutics for lung cancer therapy is still in its infancy and faces greater than expected challenges. This article focuses on the current inhalable nanocarrier-based drugs for lung cancer treatment.
Collapse
|
26
|
Zhao C, Xing Z, Zhang C, Fan Y, Liu H. Nanopharmaceutical-based regenerative medicine: a promising therapeutic strategy for spinal cord injury. J Mater Chem B 2021; 9:2367-2383. [PMID: 33662083 DOI: 10.1039/d0tb02740e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a neurological disorder that can lead to loss of perceptive and athletic function due to the severe nerve damage. To date, pieces of evidence detailing the precise pathological mechanisms in SCI are still unclear. Therefore, drug therapy cannot effectively alleviate the SCI symptoms and faces the limitations of systemic administration with large side effects. Thus, the development of SCI treatment strategies is urgent and valuable. Due to the application of nanotechnology in pharmaceutical research, nanopharmaceutical-based regenerative medicine will bring colossal development space for clinical medicine. These nanopharmaceuticals (i.e. nanocrystalline drugs and nanocarrier drugs) are designed using different types of materials or bioactive molecules, so as to improve the therapeutic effects, reduce side effects, and subtly deliver drugs, etc. Currently, an increasing number of nanopharmaceutical products have been approved by drug regulatory agencies, which has also prompted more researchers to focus on the potential treatment strategies of SCI. Therefore, the purpose of this review is to summarize and elaborate the research progress as well as the challenges and future of nanopharmaceuticals in the treatment of SCI, aiming to promote further research of nanopharmaceuticals in SCI.
Collapse
Affiliation(s)
- Chen Zhao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, P. R. China. and School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, P. R. China
| | - Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, P. R. China.
| | - Chunchen Zhang
- Key Laboratory for Biomedical Engineering of Education Ministry of China, Zhejiang University, Hangzhou, 310027, P. R. China and Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, P. R. China.
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, P. R. China.
| |
Collapse
|
27
|
Gardouh AR, Ewedah TM, Abd-Allah FI, Ghorab MM, Omran MM, El-Sawy HS. Enhanced efficacy, cellular uptake, and antiangiogenic activity of the optimized imatinib mesylate-loaded proniosomal-derived nanovesicles. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Prabhu P. Tumoral delivery of nanotherapeutics. HANDBOOK ON NANOBIOMATERIALS FOR THERAPEUTICS AND DIAGNOSTIC APPLICATIONS 2021:53-101. [DOI: 10.1016/b978-0-12-821013-0.00024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
29
|
Yan D, Wei H, Lai X, Ge Y, Xu S, Meng J, Wen T, Liu J, Zhang W, Wang J, Xu H. Co-delivery of homoharringtonine and doxorubicin boosts therapeutic efficacy of refractory acute myeloid leukemia. J Control Release 2020; 327:766-778. [DOI: 10.1016/j.jconrel.2020.09.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022]
|
30
|
Ghosh S, Lalani R, Maiti K, Banerjee S, Patel V, Bhowmick S, Misra A. Optimization and efficacy study of synergistic vincristine coloaded liposomal doxorubicin against breast and lung cancer. Nanomedicine (Lond) 2020; 15:2585-2607. [DOI: 10.2217/nnm-2020-0169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim: To improve the efficacy of poly-ethylene glycol (PEG)ylated liposomes coloaded with doxorubicin and vincristine against triple-negative breast cancer (TNBC) and non-small-cell lung cancer (NSCLC). Methods: The combinatorial index of the drugs was established using the Chou-Talalay method in MDA-MB-231 and A549 cell lines. The most effective ratio was co-encapsulated in factorial design optimized nanoliposomes which were characterized for similarity to clinical standard and evaluated in vitro and in vivo for therapeutic efficacy. Results & conclusion: The formulation exhibited more than 95% co-encapsulation, a size of 95.74 ± 2.65 nm and zeta potential of -9.17 ± 1.19 mV while having no significant differences in physicochemical and biochemical characteristics as compared with the clinical standard. Efficacy evaluation studies showed significantly improved cytotoxicity and tumor regression compared with liposomal doxorubicin indicating improvement in efficacy against TNBC and NSCLC.
Collapse
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
| | - Kuntal Maiti
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Shubhadeep Banerjee
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Formulation Research & Development Department, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat-390012, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat - 390001, India
- Pharmaceutical Research, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University, VL Mehta Road, Vile Parle (W), Mumbai, Maharashtra - 400 056, Maharashtra India
| |
Collapse
|
31
|
Ghosh S, Lalani R, Maiti K, Banerjee S, Bhatt H, Bobde YS, Patel V, Biswas S, Bhowmick S, Misra A. Synergistic co-loading of vincristine improved chemotherapeutic potential of pegylated liposomal doxorubicin against triple negative breast cancer and non-small cell lung cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102320. [PMID: 33075540 DOI: 10.1016/j.nano.2020.102320] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/06/2020] [Accepted: 10/06/2020] [Indexed: 11/15/2022]
Abstract
The current work aims to explore the biological characteristics of vincristine synergistic co-loading into pegylated liposomal doxorubicin in non-indicated modalities of non-small cell lung cancer (NSCLC) and triple negative breast cancer (TNBC). The combinatorial liposome prepared by active co-loading of the drugs against modified ammonium ion gradient exhibited 95% encapsulation of both drugs. The cellular uptake studies using confocal microscopy and flow cytometry showed significantly increased uptake of dual drug formulation as against liposomal doxorubicin. The co-loaded liposome formulation had significantly increased cell cycle arrest in G2/M phase with subsequent apoptosis and reduced cell viability in both tumor cell lines than doxorubicin liposome. This carrier exhibited similar acute toxicity, pharmacokinetic and tissue distribution profiles with significant increase in tumor regression as compared to liposomal doxorubicin. These results indicate that co-encapsulation of vincristine into clinically used pegylated liposomal doxorubicin significantly improved in-vitro and in-vivo therapeutic efficacy against NSCLC and TNBC.
Collapse
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Kuntal Maiti
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Shubhadeep Banerjee
- Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Himanshu Bhatt
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Yamini Shankar Bobde
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India.
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Pharmaceutical Research, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University, Mumbai, Maharashtra, India.
| |
Collapse
|
32
|
Nanocarriers in effective pulmonary delivery of siRNA: current approaches and challenges. Ther Deliv 2020; 10:311-332. [PMID: 31116099 DOI: 10.4155/tde-2019-0012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Research on siRNA is increasing due to its wide applicability as a therapeutic agent in irreversible medical conditions. siRNA inhibits expression of the specific gene after its delivery from formulation to cytosol region of a cell. RNAi (RNA interference) is a mechanism by which siRNA is silencing gene expression for a particular disease. Numerous studies revealed that naked siRNA delivery is not preferred due to instability and poor pharmacokinetic performance. Nanocarriers based delivery of siRNA has the advantage to overcome physiological barriers and protect the integrity of siRNA from degradation by RNAase. Various diseases like lung cancer, cystic fibrosis, asthma, etc can be treated effectively by local lung delivery. The selective targeted therapeutic action in diseased organ and least off targeted cytotoxicity are the key benefits of pulmonary delivery. The current review highlights recent developments in pulmonary delivery of siRNA with novel nanosized formulation approach with the proven in vitro/in vivo applications.
Collapse
|
33
|
Li Y, Lin J, Cai Z, Wang P, Luo Q, Yao C, Zhang Y, Hou Z, Liu J, Liu X. Tumor microenvironment-activated self-recognizing nanodrug through directly tailored assembly of small-molecules for targeted synergistic chemotherapy. J Control Release 2020; 321:222-235. [PMID: 32061620 DOI: 10.1016/j.jconrel.2020.02.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 12/16/2022]
Abstract
Carrier-free nanodrug via small-molecule assembly is a promising alternative strategy for tumor therapy. Thus, developing a self-recognizing carrier-free nanodrug without introduction of foreign ligand is very attractive to meet both targeting and therapeutic requirements while reducing structural complexity. Here we fabricated a tumor microenvironment-activated self-targeting nanodrug, via co-assembly of hydroxycamptothecin (HCPT) and bi-functional methotrexate (MTX, not only has antitumor effect but also shows innate affinity towards folate receptors) followed by surface covering through acidity-responsive polyethylene glycol (PEG). Notably, the morphology and size of MTX-HCPT nanodrug could be tuned by varying the drug-to-drug ratio and assembly time. The PEG shell of our nanodrug could be detached in response to acidic tumor microenvironment, and then MTX could be exposed for self-targeting to enhance tumor cell uptake. Subsequently, the shell-detached nanodrug could be dissociated in relatively stronger acidic lysosomal environment, resulting in burst release of both drugs. Further in vitro and in vivo studies demonstrated that our nanodrug showed a ~2.98-fold increase in cancer cell uptake, a ~1.25-fold increase in drug accumulation at tumor site, a significantly lower CI50 value of ~0.3, a ~27.3% improvement in tumor inhibition comparing with the corresponding non-responsive nanodrug. Taken together, the here reported tumor microenvironment-activated self-recognizing nanodrug might be an extremely promising strategy for synergistically enhancing chemotherapy efficiency with minimized side effects.
Collapse
Affiliation(s)
- Yang Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China
| | - Jinyan Lin
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Peiyuan Wang
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China
| | - Qiang Luo
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China
| | - Cuiping Yao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yun Zhang
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China
| | - Zhenqing Hou
- College of Materials, Research Center of Biomedical Engineering of Xiamen & Key Laboratory of Biomedical Engineering of Fujian Province, Xiamen University, Xiamen 361005, PR China.
| | - Jingfeng Liu
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China.
| | - Xiaolong Liu
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, PR China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, PR China.
| |
Collapse
|
34
|
Ma W, Sun J, Xu J, Luo Z, Diao D, Zhang Z, Oberly PJ, Minnigh MB, Xie W, Poloyac SM, Huang Y, Li S. Sensitizing Triple Negative Breast Cancer to Tamoxifen Chemotherapy via a Redox-Responsive Vorinostat-containing Polymeric Prodrug Nanocarrier. Am J Cancer Res 2020; 10:2463-2478. [PMID: 32194813 PMCID: PMC7052901 DOI: 10.7150/thno.38973] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/15/2019] [Indexed: 01/07/2023] Open
Abstract
There is an urgent and unmet need to develop effective therapies for triple negative breast cancers (TNBCs) which are much more aggressive and have poor prognosis due to lack of receptor targets for Her2-targeted and endocrine therapy. In this study we systematically evaluated the effect of Vorinostat (SAHA, a pan-HDAC inhibitor) in reactivating the expression of functional estrogen receptor α (ERα) and synergizing with tamoxifen (TAM, a selective estrogen-receptor modulator) in antitumor activity. In addition, a SAHA prodrug-based dual functional nanocarrier was developed for codelivery of SAHA and TAM for effective combination therapy. Methods: A SAHA-containing polymeric nanocarrier, POEG-co-PVDSAHA was developed via reversible addition-fragmentation transfer (RAFT) polymerization with SAHA incorporated into the polymer through a redox-responsive disulfide linkage. The effect of both free SAHA and POEG-co-PVDSAHA on reactivating the expression of functional ERα was investigated in several human and murine TNBC cell lines via examining the mRNA and protein expression of ERα target genes. The cytotoxicity of free SAHA and TAM combination and TAM-loaded POEG-co-PVDSAHA micelles was examined via MTT assay. The in vivo antitumor activity of TAM-loaded POEG-co-PVDSAHA was investigated in a murine breast cancer model (4T1.2). Results: Both free SAHA and POEG-co-PVDSAHA were effective in inducing the reexpression of functional estrogen receptor α (ERα), which may have helped to sensitize TNBCs to TAM. More importantly, POEG-co-PVDSAHA self-assembled to form small-sized micellar carrier that is effective in formulating and codelivery of TAM. TAM-loaded POEG-co-PVDSAHA micelles exhibited enhanced and synergistic cytotoxicity against TNBC cell lines compared with free SAHA, free TAM and TAM loaded into a pharmacologically inert control carrier (POEG-co-PVMA). In addition, codelivery of TAM via POEG-co-PVDSAHA micelles led to significantly improved antitumor efficacy in 4T1.2 tumor model compared with other groups such as combination of free SAHA and TAM and TAM-loaded POEG-co-PVMA micelles. Conclusion: Our prodrug-based co-delivery system may provide an effective and simple strategy to re-sensitize TNBCs to TAM-based hormone therapy.
Collapse
|
35
|
Ghosh S, Lalani R, Patel V, Bhowmick S, Misra A. Surface engineered liposomal delivery of therapeutics across the blood brain barrier: recent advances, challenges and opportunities. Expert Opin Drug Deliv 2019; 16:1287-1311. [DOI: 10.1080/17425247.2019.1676721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
36
|
Xiang S, Zhang K, Yang G, Gao D, Zeng C, He M. Mitochondria-Targeted and Resveratrol-Loaded Dual-Function Titanium Disulfide Nanosheets for Photothermal-Triggered Tumor Chemotherapy. NANOSCALE RESEARCH LETTERS 2019; 14:211. [PMID: 31227943 PMCID: PMC6588667 DOI: 10.1186/s11671-019-3044-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/07/2019] [Indexed: 05/04/2023]
Abstract
A subcellular organelle-targeted delivery of anti-cancer drugs is a promising strategy to maximize the anti-cancer effects and minimize the adverse effects. Herein, we prepared a mitochondria-targeted drug delivery nanoplatform based on IR780 iodide (IR780) and titanium disulfide (TiS2) nanosheets. Due to the large specific surface area of TiS2 nanosheets, the nanoplatform could highly load anti-cancer drug resveratrol (RV). The as-prepared nanocomposite (IR780-TiS2/RV) was used for an efficacious photothermal-triggered tumor chemotherapy. IR780-TiS2/RV showed satisfactory stability and biocompatibility, and the loading ratio of RV and IR780 was about 112% and 56%, respectively. Upon the near-infrared (NIR) irradiation, the heat generated by IR780-TiS2/RV could trigger the RV release. Due to the conjugation with the mitochondria-specific IR780, IR780-TiS2/RV could target and accumulate in mitochondria and release RV when triggered by NIR to decrease the mitochondrial membrane potential, rapidly induce the upregulation of key intrinsic apoptotic factors such as cytochrome c, and initiate the caspase cascade, thereby achieving the chemotherapeutic effect. The IR780-TiS2/RV nanocomposite was demonstrated to have a high anti-tumor efficacy in vitro and in vivo as well as no remarkable tissue toxicity. We believe our study demonstrates that the NIR-triggered IR780-TiS2/RV nanoplatform could be a promising chemotherapeutic agent in clinical practice.
Collapse
Affiliation(s)
- Sen Xiang
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| | - Kaifang Zhang
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| | - Guanghua Yang
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| | - Dongdong Gao
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| | - Chen Zeng
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| | - Miao He
- The First Department of Oncology, Zhumadian Central Hospital, 747 Zhumadian Zhonghua Road, Zhumadian, 463000 China
| |
Collapse
|
37
|
Lim C, Won WR, Moon J, Sim T, Shin Y, Kim JC, Lee ES, Youn YS, Oh KT. Co-delivery of d-(KLAKLAK)2 peptide and doxorubicin using a pH-sensitive nanocarrier for synergistic anticancer treatment. J Mater Chem B 2019. [DOI: 10.1039/c9tb00741e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Currently, one of the most important challenges in the development of nanotechnology-based anticancer treatments is the failure of nanoparticles to escape from the endo-lysosomal compartment.
Collapse
Affiliation(s)
- Chaemin Lim
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Woong Roeck Won
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Junseong Moon
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Taehoon Sim
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Yuseon Shin
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Jae Chang Kim
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| | - Eun Seong Lee
- Division of Biotechnology
- The Catholic University of Korea
- Bucheon 14662
- South Korea
| | - Yu Seok Youn
- Department of Pharmaceutical Sciences
- School of Pharmacy
- Sungkyunkwan University
- Suwon 16419
- South Korea
| | - Kyung Taek Oh
- Department of Pharmaceutical Sciences
- College of Pharmacy
- Chung-Ang University
- Seoul 06974
- South Korea
| |
Collapse
|