1
|
Yadav PK, Chauhan D, Yadav P, Tiwari AK, Sultana N, Gupta D, Mishra K, Gayen JR, Wahajuddin M, Chourasia MK. Nanotechnology Assisted Drug Delivery Strategies for Chemotherapy: Recent Advances and Future Prospects. ACS APPLIED BIO MATERIALS 2025; 8:3601-3622. [PMID: 40318022 DOI: 10.1021/acsabm.5c00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
In pursuit of the treatment of cancer, nanotechnology engineering has emerged as the simplest and most effective means, with the potential to deliver antitumor chemotherapeutics at the targeted site. Employing nanotechnology for drug delivery provides diverse nanosize particles ranging from one to a thousand nanometers. Reduced size improves drug bioavailability by increasing drug diffusion and decreasing the efflux rate. These nanocarriers offer an enormous scope for modification following the chemical and biological properties of both the drug and its disease. Moreover, these nanoformulations assist in targeting pharmaceutically active drug molecules to the desired site and have gained importance in recent years. Their modern use has revolutionized the antitumor action of many therapeutic agents. Higher drug loading efficiency, thermal stability, easy fabrication, low production cost, and large-scale industrial production draw attention to the application of nanotechnology as a better platform for the delivery of drug molecules. Furthermore, the interaction of nanocarrier technology-assisted agents lowers a drug's toxicity and therapeutic dosage, reduces drug tolerance, and enhances active drug concentration in neoplasm tissue, thus decreasing the concentration in healthy tissue. Nanotechnology-based medications are being widely explored and have depicted effective cancer management in vivo and in vitro systems, leading to many clinical trials with promising results. This review summarizes the innovative impact and application of different nanocarriers developed in recent years in cancer therapy. Subsequently, it also describes the essential findings and methodologies and their effects on cancer treatment. Compared with conventional therapy, nanomedicines can significantly improve the therapeutic effectiveness of antitumor drugs. Thus, the adverse effects associated with healthy tissues are decreased, and adverse effects are scaled back through enhanced permeability and retention effects. Lastly, future insights assisting nanotechnology in active therapeutics delivery and their scope in cancer chemotherapeutics have also been discussed.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Chauhan
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nazneen Sultana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Deepak Gupta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Keerti Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Tiwari A, Yadav P, Shah AA, Rana R, Yadav P, Mishra K, Tripathi S, Kothuri N, Verma S, Kashyap A, Jaiswal S, Verma S, Mugale MN, Chourasia MK. Designing pegylated dextran sheathed doxorubicin loaded iron nanoparticles against premenopausal breast cancer. Int J Biol Macromol 2025; 307:141874. [PMID: 40064263 DOI: 10.1016/j.ijbiomac.2025.141874] [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: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
Premenopausal women, often iron-deficient, face a heightened risk of breast cancer. Magnetic nanoparticles (MNPs) show promise for cancer therapy but are limited by challenges in pharmacokinetics, biocompatibility, and magnetic property stability, leading to reduced efficacy and resistance. To overcome these hurdles, a double-shelled magnetic nanoparticle (DOX-RA-MNP) system was developed for pH-sensitive delivery of Retinoic acid and Doxorubicin using an immunomodulatory polymeric approach. Optimized by using a QbD framework, the formulation demonstrated ideal size, polydispersity index, zeta potential, and enhanced doxorubicin loading. The formulation depicted sustained drug release with enhanced release at tumor pH. In vitro studies on MDA-MB-231 cells revealed improved cytotoxicity, cellular uptake, G2 phase cell cycle arrest, mitochondrial membrane depolarization, and PgP protein inhibition. In in vivo, the system showed significant tumor regression, favorable pharmacokinetics, biodistribution, and safety, with lower hemolysis and improved survival rates. The biochemical studies provide insights about the role of ferroptosis increasing reactive oxygen species (ROS) level and immunomodulatory effects. Further, the lower hemolysis and enhanced survival of animals confirmed safety of the developed formulation. These findings suggest the DOX-RA-MNP system effectively targets and localizes drugs, reducing toxicity and offering a potent strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Amrendra Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Pavan Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Aarti Abhishek Shah
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Rafquat Rana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Keerti Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shourya Tripathi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naresh Kothuri
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sonia Verma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit Kashyap
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Srishty Jaiswal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shobhit Verma
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Madhav N Mugale
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
3
|
Wang C, Xiu Y, Zhang Y, Wang Y, Xu J, Yu W, Xing D. Recent advances in biotin-based therapeutic agents for cancer therapy. NANOSCALE 2025; 17:1812-1873. [PMID: 39676680 DOI: 10.1039/d4nr03729d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Biotin receptors, as biomarkers for cancer cells, are overexpressed in various tumor types. Compared to other vitamin receptors, such as folate receptors and vitamin B12 receptors, biotin receptor-based targeting strategies exhibit superior specificity and broader potential in treating aggressive cancers, including ovarian cancer, leukemia, colon cancer, breast cancer, kidney cancer, and lung cancer. These strategies promote biotin transport via receptor-mediated endocytosis, which is triggered upon ligand binding. Biotin, as the ligand of the biotin receptor, can be conjugated to anti-cancer drugs to form targeted therapies that bind to receptors overexpressed on tumor cells, thus increasing drug uptake. Despite these advantages, many candidate drugs have progressed slowly and remain in the preclinical stage, impeding clinical translation. This is mainly due to the effects of various conjugation methods and drug formulations on their functionality and efficacy. Therefore, developing novel biotin-based therapeutics is crucial. The innovation of this strategy lies in its multifunctionality-researchers can use different conjugation methods to design and synthesize these drugs, enabling precise targeting of various tumor types while minimizing toxicity to normal cells. These drugs include small-molecule-biotin conjugates (SMBCs) and nano-biotin conjugates (NBCs). This dual-platform approach represents a significant advancement in targeted therapy, offering unprecedented flexibility in drug design and delivery. Compared to chemotherapy drugs and traditional delivery systems, biotin-based drugs with tumor-specific targeting demonstrate enhanced targeting, improved efficacy, and reduced toxicity. This review examines strategies and applications for enhancing the delivery of chemotherapy drugs to cancer cells, highlighting the need for high-quality conjugates and strategies. It not only summarizes the latest progress but also provides key insights into how this emerging field could revolutionize personalized cancer treatment, especially in the context of precision medicine. Additionally, it offers perspectives on future research directions in this field.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yutao Xiu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Yanhong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
4
|
Lee DJ, Cao Y, Juvekar V, Sauraj, Noh CK, Shin SJ, Liu Z, Kim HM. Development of a small molecule-based two-photon photosensitizer for targeting cancer cells. J Mater Chem B 2024. [PMID: 39469993 DOI: 10.1039/d4tb01706d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Photodynamic therapy (PDT) employing two-photon (TP) excitation is increasingly recognized to induce cell damage selectively in targeted areas, underscoring the importance of developing TP photosensitizers (TP-PSs). In this study, we developed BSe-B, a novel PS that combines a selenium containing dye with biotin, a cancer-selective ligand, and is optimized for TP excitation. BSe-B demonstrated enhanced cancer selectivity, efficient generation of type-I based reactive oxygen species (ROS), low dark toxicity, and excellent cell-staining capability. Evaluation across diverse cell lines (HeLa, A549, OVCAR-3, WI-38, and L-929) demonstrated that BSe-B differentiated and targeted cancer cells while sparing normal cells. BSe-B displayed excellent in vivo biocompatibility. In cancer models such as three-dimensional spheroids and actual colon cancer tissues, BSe-B selectively induced ROS production and cell death under TP irradiation, demonstrating precise spatial control. These findings highlight the potential of BSe-B for imaging-guided PDT and its capability for micro treatment within tissues. Thus, BSe-B demonstrates robust TP-PDT capabilities, making it a promising dual-purpose tool for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dong Joon Lee
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Yu Cao
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Vinayak Juvekar
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Sauraj
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Zhihong Liu
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Hwan Myung Kim
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
5
|
Mhaske A, Kaur J, Naqvi S, Shukla R. Decitabine enclosed biotin-zein conjugated nanoparticles: synthesis, characterization, in vitro and in vivo evaluation. Nanomedicine (Lond) 2024; 19:1743-1760. [PMID: 39041671 PMCID: PMC11418219 DOI: 10.1080/17435889.2024.2374700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 06/27/2024] [Indexed: 07/24/2024] Open
Abstract
Aim: This study focuses on biotinylated nanocarriers designed to encapsulate amphiphilic molecules with self-biodegradable properties for enhanced drug delivery.Methods: Biotin-zein conjugated nanoparticles were synthesized and tested in C6 cell lines to evaluate their viability and cellular uptake. Optimization was achieved using a a central composite design. The nanoparticles underwent thermogravimetric analysis, and their pharmacokinetics and biodistribution were also studied.Results: The optimized nanoparticles displayed 96.31% drug encapsulation efficiency, a particle size of 95.29 nm and a zeta potential of -17.7 mV. These nanoparticles showed increased cytotoxicity and improved cellular uptake compared with free drugs. Thermogravimetric analysis revealed that the drug-loaded nanocarriers provided better protection against drug degradation. Pharmacokinetic and biodistribution studies indicated that the formulation had an extended brain residence time, highlighting its effectiveness.Conclusion: The biotin-zein conjugated nanoparticles developed in this study offer a promising nano-vehicle for in vivo biodistribution and pharmacokinetic applications. Their high drug encapsulation efficiency, stability and extended brain residence time suggest they are effective for targeted drug delivery and therapeutic uses.
Collapse
Affiliation(s)
- Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Jasleen Kaur
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Saba Naqvi
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
- Department of Pharmacology & Regulatory Toxicology, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, 226002, India
| |
Collapse
|
6
|
Jin S, Huang J, Dong Y, Hu D, Sun J, Li Z, Zhong BY, Liu Z, Zhu R, Wang G. Ultrastable PLGA-Coated 177Lu-Microspheres for Radioembolization Therapy of Hepatocellular Carcinoma. Mol Pharm 2024; 21:3407-3415. [PMID: 38822792 DOI: 10.1021/acs.molpharmaceut.4c00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
Transarterial radioembolization (TARE) is a highly effective localized radionuclide therapy that has been successfully used to treat hepatocellular carcinoma (HCC). Extensive research has been conducted on the use of radioactive microspheres (MSs) in TARE, and the development of ideal radioactive MSs is crucial for clinical trials and patient treatment. This study presents the development of a radioactive MS for TARE of HCC. These MSs, referred to as 177Lu-MS@PLGA, consist of poly(lactic-co-glycolic acid) (PLGA) copolymer and radioactive silica MSs, labeled with 177Lu and then coated with PLGA. It has an extremely high level of radiostability. Cellular experiments have shown that it can cause DNA double-strand breaks, leading to cell death. In vivo radiostability of 177Lu-MS@PLGA is demonstrated by microSPECT/CT imaging. In addition, the antitumor study has shown that TARE of 177Lu-MS@PLGA can effectively restrain tumor growth without harmful side effects. Thus, 177Lu-MS@PLGA exhibits significant potential as a radioactive MS for the treatment of HCC.
Collapse
Affiliation(s)
- Shuo Jin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jintao Huang
- Department of Interventional Radiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yi Dong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Di Hu
- Department of Interventional Radiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jing Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Zhihao Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Bin-Yan Zhong
- Department of Interventional Radiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhiyong Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ran Zhu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
7
|
Saklani R, Yadav PK, Tiwari AK, Gawali SL, Hassan PA, Yadav K, Mugale MN, Kalleti N, Rath SK, Mishra DP, Dierking I, Chourasia MK. Synchronized Codelivery of Combination Chemotherapies Intratumorally Using a Lipidic Lyotropic Liquid Crystal System. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29098-29111. [PMID: 38780083 DOI: 10.1021/acsami.4c01432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
In this work, an injectable in situ depot-forming lipidic lyotropic liquid crystal (L3C) system is developed to codeliver a precisely synchronized combination of chemotherapeutics intratumorally. The developed L3C system is composed of amphiphilic lipids and surfactants, including monoolein, phosphatidylcholine, tocopherol acetate, and d-α-tocopherol polyethylene glycol 1000 succinate. Owing to its amphiphilic nature, the developed formulation can coaccommodate both hydrophobic and hydrophilic chemotherapeutic moieties simultaneously. The study presents a proof of concept by designing a combination chemotherapy regimen in vitro and demonstrating its in vivo translation using doxorubicin and paclitaxel as model hydrophilic and hydrophobic drug moieties, respectively. The synchronized combination of the two chemotherapeutics with maximum synergistic activity was identified, coloaded in the developed L3C system at predefined stoichiometric ratios, and evaluated for antitumor efficacy in the 4T1 breast tumor model in BALB/c mice. The drug-loaded L3C formulation is a low-viscosity injectable fluid with a lamellar phase that transforms into a hexagonal mesophase depot system upon intratumoral injection. The drug-loaded depot system locally provides sustained intratumoral delivery of the chemotherapeutics combination at their precisely synchronized ratio for over a period of one month. Results demonstrate that the exposure of the tumor to the precisely synchronized intratumoral chemotherapeutics combination via the developed L3C system resulted in significantly higher antitumor activity and reduced cardiotoxicity compared to the unsynchronized combination chemotherapy or the synchronized but uncoordinated drug delivery administered by a conventional intravenous route. These findings demonstrate the potential of the developed L3C system for achieving synchronized codelivery of the chemotherapeutics combination intratumorally and improving the efficacy of combination chemotherapy.
Collapse
Affiliation(s)
- Ravi Saklani
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh L Gawali
- Nanotherapeutics and Biosensors Section, Chemistry Division, Bhabha Atomic Research Centre Trombay, Mumbai 400085, India
| | - Puthusserickal A Hassan
- Nanotherapeutics and Biosensors Section, Chemistry Division, Bhabha Atomic Research Centre Trombay, Mumbai 400085, India
| | - Karan Yadav
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031 India
| | - Madhav N Mugale
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031 India
| | - Navodayam Kalleti
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031 India
| | - Srikanta K Rath
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031 India
| | - Durga P Mishra
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ingo Dierking
- Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
8
|
Chen MM, Tang X, Li JJ, Chen FY, Jiang ZT, Fu R, Li HB, Hu XY, Geng WC, Guo DS. Active targeting tumor therapy using host-guest drug delivery system based on biotin functionalized azocalix[4]arene. J Control Release 2024; 368:691-702. [PMID: 38492860 DOI: 10.1016/j.jconrel.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Host-guest drug delivery systems (HGDDSs) provided a facile method for incorporating biomedical functions, including efficient drug-loading, passive targeting, and controlled drug release. However, developing HGDDSs with active targeting is hindered by the difficult functionalization of popular macrocycles. Herein, we report an active targeting HGDDS based on biotin-modified sulfonated azocalix[4]arene (Biotin-SAC4A) to efficiently deliver drug into cancer cells for improving anti-tumor effect. Biotin-SAC4A was synthesized by amide condensation and azo coupling. Biotin-SAC4A demonstrated hypoxia responsive targeting and active targeting through azo and biotin groups, respectively. DOX@Biotin-SAC4A, which was prepared by loading doxorubicin (DOX) in Biotin-SAC4A, was evaluated for tumor targeting and therapy in vitro and in vivo. DOX@Biotin-SAC4A formulation effectively killed cancer cells in vitro and more efficiently delivered DOX to the lesion than the similar formulation without active targeting. Therefore, DOX@Biotin-SAC4A significantly improved the in vivo anti-tumor effect of free DOX. The facilely prepared Biotin-SAC4A offers strong DOX complexation, active targeting, and hypoxia-triggered release, providing a favorable host for effective breast cancer chemotherapy in HGDDSs. Moreover, Biotin-SAC4A also has potential to deliver agents for other therapeutic modalities and diseases.
Collapse
Affiliation(s)
- Meng-Meng Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Xingchen Tang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Juan-Juan Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Fang-Yuan Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Rong Fu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Hua-Bin Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China.
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Collaborative Innovation Center of Chemical Science and Engineering, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
9
|
Giram P, Nimma R, Bulbule A, Yadav AS, Gorain M, Venkata Radharani NN, Kundu GC, Garnaik B. Poly(d,l-lactide- co-glycolide) Surface-Anchored Biotin-Loaded Irinotecan Nanoparticles for Active Targeting of Colon Cancer. ACS OMEGA 2024; 9:3807-3826. [PMID: 38284072 PMCID: PMC10809773 DOI: 10.1021/acsomega.3c07833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024]
Abstract
A poly(d,l-lactide-co-glycolide) (PLGA) copolymer was synthesized using the ring-opening polymerization of d,l-lactide and glycolide monomers in the presence of zinc proline complex in bulk through the green route and was well characterized using attenuated total reflectance-Fourier transform infrared, 1H and 13C nuclear magnetic resonance, gel permeation chromatography, differential scanning calorimetry, X-ray diffraction, matrix-assisted laser desorption/ionization time-of-flight, etc. Furthermore, PLGA-conjugated biotin (PLGA-B) was synthesized using the synthesized PLGA and was employed to fabricate nanoparticles for irinotecan (Ir) delivery. These nanoparticles (PLGA-NP-Ir and PLGA-B-NP-Ir) were tested for physicochemical and biological characteristics. PLGA-B-NP-Ir exhibited a stronger cellular uptake and anticancer activity as compared to PLGA-NP-Ir in CT-26 cancer cells (log p < 0.05). The accumulation and retention of fluorescence-labeled nanoparticles were observed to be better in CT-26-inoculated solid tumors in Balb/c mice. The PLGA-B-NP-Ir-treated group inhibited tumor growth significantly more (log p < 0.001) than the untreated control, PLGA-NP-Ir, and Ir-treated groups. Furthermore, no body weight loss, hematological, and blood biochemical tests demonstrated the nanocarriers' nontoxic nature. This work presents the use of safe PLGA and the demonstration of a proof-of-concept of biotin surface attached PLGA nanoparticle-mediated active targeted Ir administration to combat colon cancer. To treat colon cancer, PLGA-B-NP-Ir performed better due to specific active tumor targeting and greater cellular uptake due to biotin.
Collapse
Affiliation(s)
- Prabhanjan
S. Giram
- Polymer
Science and Engineering Division, CSIR-National
Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy
of Scientific and Innovative Research AcSIR Headquarters, CSIR-HRDC Campus Sector 19, Kamla
Nehru Nagar, Ghaziabad, Uttar
Pradesh 201 002, India
| | - Ramakrishna Nimma
- Laboratory
of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Anuradha Bulbule
- Laboratory
of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Amit Singh Yadav
- Laboratory
of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | - Mahadeo Gorain
- Laboratory
of Tumor, Biology, Angiogenesis and Nanomedicine Research, National Center for Cell Science, Pune 411007, India
| | | | - Gopal C. Kundu
- School
of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Institute of Eminence, Bhubaneswar 751 024, India
| | - Baijayantimala Garnaik
- Polymer
Science and Engineering Division, CSIR-National
Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy
of Scientific and Innovative Research AcSIR Headquarters, CSIR-HRDC Campus Sector 19, Kamla
Nehru Nagar, Ghaziabad, Uttar
Pradesh 201 002, India
| |
Collapse
|
10
|
Yadav PK, Saklani R, Tiwari AK, Verma S, Chauhan D, Yadav P, Rana R, Kalleti N, Gayen JR, Wahajuddin, Rath SK, Mugale MN, Mitra K, Chourasia MK. Ratiometric codelivery of Paclitaxel and Baicalein loaded nanoemulsion for enhancement of breast cancer treatment. Int J Pharm 2023; 643:123209. [PMID: 37422142 DOI: 10.1016/j.ijpharm.2023.123209] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
The most prevalent clinical option for treating cancer is combination chemotherapy. In combination therapy, assessment and optimization for obtaining a synergistic ratio could be obtained by various preclinical setups. Currently, in vitro optimization is used to get synergistic cytotoxicity while constructing combinations. Herein, we co-encapsulated Paclitaxel (PTX) and Baicalein (BCLN) with TPP-TPGS1000 containing nanoemulsion (TPP-TPGS1000-PTX-BCLN-NE) for breast cancer treatment. The assessment of cytotoxicity of PTX and BCLN at different molar weight ratios provided an optimized synergistic ratio (1:5). Quality by Design (QbD) approach was later applied for the optimization as well as characterization of nanoformulation for its droplet size, zeta potential and drug content. TPP-TPGS1000-PTX-BCLN-NE significantly enhanced cellular ROS, cell cycle arrest, and depolarization of mitochondrial membrane potential in the 4T1 breast cancer cell line compared to other treatments. In the syngeneic 4T1 BALB/c tumor model, TPP-TPGS1000-PTX-BCLN-NE outperformed other nanoformulation treatments. The pharmacokinetic, biodistribution and live imaging studies pivoted TPP-TPGS1000-PTX-BCLN-NE enhanced bioavailability and PTX accumulation at tumor site. Later, histology studies confirmed nanoemulsion non-toxicity, expressing new opportunities and potential to treat breast cancer. These results suggested that current nanoformulation can be a potential therapeutic approach to effectively address breast cancer therapy.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ravi Saklani
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Saurabh Verma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Divya Chauhan
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Rafquat Rana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Navodayam Kalleti
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Wahajuddin
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Srikanta K Rath
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Madhav N Mugale
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Kalyan Mitra
- Electron Microscopy Division, Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India.
| |
Collapse
|
11
|
Elkodous MA, Olojede SO, Sahoo S, Kumar R. Recent advances in modification of novel carbon-based composites: Synthesis, properties, and biotechnological/ biomedical applications. Chem Biol Interact 2023; 379:110517. [PMID: 37149208 DOI: 10.1016/j.cbi.2023.110517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 03/12/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
Nowadays, carbon-based materials owing to great interest in biomedical science/biotechnology and applied for effective diagnosis and treatment of disease. To enhance the effectiveness of carbon nanotubes (CNTs)/graphene-based materials for bio-medical science/technology applications, different kinds of surface modification/functionalization were developed for the attachment of metal oxides nanostructures, biomolecules and polymers. The attachment of pharmaceutical agents with CNTs/graphene, make it a favorable candidate in research field of bio-medical science/technology applications. Surface modified/functionalized CNTs and graphene derivatives materials integrated with pharmaceutical agents has been developed for the purpose of cancer therapy, antibacterial action, pathogens bio detection, drug and gene delivery. Surface modification or functionalization of CNT/graphene materials provides good platform for pharmaceutical agents attachment with improved surface Raman scattering, fluorescence and its quenching capability. Graphene-based biosensing and bioimaging technologies are widely applied to identify numerous trace level analytes. These fluorescent and electrochemical sensors are utilized primarily for detecting organic, inorganic, and biomolecules. In this article, we highlights and summarized overview of the current research progress concerned on the CNTs/graphene-based materials as a new generation materials for detection and treatment of diseases.
Collapse
Affiliation(s)
- M Abd Elkodous
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi, 441-8580, Japan; Center for Nanotechnology (CNT), School of Engineering and Applied Sciences, Nile University, Sheikh Zayed, Giza, 16453, Egypt
| | - Samuel Oluwaseun Olojede
- Nanotechnology Platforms, Discipline of Clinical Anatomy, School of Laboratory Medicine & Medical Sciences, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Sumanta Sahoo
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| | - Rajesh Kumar
- Department of Mechanical Engineering, Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India.
| |
Collapse
|
12
|
Yadav PK, Saklani R, Tiwari AK, Verma S, Rana R, Chauhan D, Yadav P, Mishra K, Kedar AS, Kalleti N, Gayen JR, Wahajuddin M, Rath SK, Mugale MN, Mitra K, Sharma D, Chourasia MK. Enhanced apoptosis and mitochondrial cell death by paclitaxel-loaded TPP-TPGS 1000-functionalized nanoemulsion. Nanomedicine (Lond) 2023; 18:343-366. [PMID: 37140535 DOI: 10.2217/nnm-2022-0268] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Background: The present research was designed to develop a nanoemulsion (NE) of triphenylphosphine-D-α-tocopheryl-polyethylene glycol succinate (TPP-TPGS1000) and paclitaxel (PTX) to effectively deliver PTX to improve breast cancer therapy. Materials & methods: A quality-by-design approach was applied for optimization and in vitro and in vivo characterization were performed. Results: The TPP-TPGS1000-PTX-NE enhanced cellular uptake, mitochondrial membrane depolarization and G2M cell cycle arrest compared with free-PTX treatment. In addition, pharmacokinetics, biodistribution and in vivo live imaging studies in tumor-bearing mice showed that TPP-TPGS1000-PTX-NE had superior performance compared with free-PTX treatment. Histological and survival investigations ascertained the nontoxicity of the nanoformulation, suggesting new opportunities and potential to treat breast cancer. Conclusion: TPP-TPGS1000-PTX-NE improved the efficacy of breast cancer treatment by enhancing its effectiveness and decreasing drug toxicity.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ravi Saklani
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Saurabh Verma
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rafquat Rana
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Divya Chauhan
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Keerti Mishra
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ashwini S Kedar
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Navodayam Kalleti
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Muhammad Wahajuddin
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Srikanta K Rath
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav N Mugale
- Division of Toxicology & Experiment Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Kalyan Mitra
- Electron Microscopy Division, Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Deepak Sharma
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Manish K Chourasia
- Division of Pharmaceutics & Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
13
|
Zhang D, Liu L, Wang J, Zhang H, Zhang Z, Xing G, Wang X, Liu M. Drug-loaded PEG-PLGA nanoparticles for cancer treatment. Front Pharmacol 2022; 13:990505. [PMID: 36059964 PMCID: PMC9437283 DOI: 10.3389/fphar.2022.990505] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022] Open
Abstract
Nanoparticles based on single-component synthetic polymers, such as poly (lactic acid-co-glycolic acid) (PLGA), have been extensively studied for antitumor drug delivery and adjuvant therapy due to their ability to encapsulate and release drugs, as well as passively target tumors. Amphiphilic block co-polymers, such as polyethylene glycol (PEG)-PLGA, have also been used to prepare multifunctional nanodrug delivery systems with prolonged circulation time and greater bioavailability that can encapsulate a wider variety of drugs, including small molecules, gene-targeting drugs, traditional Chinese medicine (TCM) and multi-target enzyme inhibitors, enhancing their antitumor effect and safety. In addition, the surface of PEG-PLGA nanoparticles has been modified with various ligands to achieve active targeting and selective accumulation of antitumor drugs in tumor cells. Modification with two ligands has also been applied with good antitumor effects, while the use of imaging agents and pH-responsive or magnetic materials has paved the way for the application of such nanoparticles in clinical diagnosis. In this work, we provide an overview of the synthesis and application of PEG-PLGA nanoparticles in cancer treatment and we discuss the recent advances in ligand modification for active tumor targeting.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Pharmaceutical Department of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hong Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Gang Xing
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xuan Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Xuan Wang, ; Minghua Liu,
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- *Correspondence: Xuan Wang, ; Minghua Liu,
| |
Collapse
|
14
|
Hartmeier PR, Kosanovich JL, Velankar KY, Armen-Luke J, Lipp MA, Gawalt ES, Giannoukakis N, Empey KM, Meng WS. Immune Cells Activating Biotin-Decorated PLGA Protein Carrier. Mol Pharm 2022; 19:2638-2650. [PMID: 35621214 PMCID: PMC10105284 DOI: 10.1021/acs.molpharmaceut.2c00343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nanoparticle formulations have long been proposed as subunit vaccine carriers owing to their ability to entrap proteins and codeliver adjuvants. Poly(lactic-co-glycolic acid) (PLGA) remains one of the most studied polymers for controlled release and nanoparticle drug delivery, and numerous studies exist proposing PLGA particles as subunit vaccine carriers. In this work we report using PLGA nanoparticles modified with biotin (bNPs) to deliver proteins via adsorption and stimulate professional antigen-presenting cells (APCs). We present evidence showing bNPs are capable of retaining proteins through the biotin-avidin interaction. Surface accessible biotin bound both biotinylated catalase (bCAT) through avidin and streptavidin horseradish peroxidase (HRP). Analysis of the HRP found that activity on the bNPs was preserved once captured on the surface of bNP. Further, bNPs were found to have self-adjuvant properties, evidenced by bNP induced IL-1β, IL-18, and IL-12 production in vitro in APCs, thereby licensing the cells to generate Th1-type helper T cell responses. Cytokine production was reduced in avidin precoated bNPs (but not with other proteins), suggesting that the proinflammatory response is due in part to exposed biotin on the surface of bNPs. bNPs injected subcutaneously were localized to draining lymph nodes detectable after 28 days and were internalized by bronchoalveolar lavage dendritic cells and macrophages in mice in a dose-dependent manner when delivered intranasally. Taken together, these data provide evidence that bNPs should be explored further as potential adjuvanting carriers for subunit vaccines.
Collapse
Affiliation(s)
- Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Jessica L Kosanovich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Jennifer Armen-Luke
- Department of Chemistry and Biochemistry, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Madeline A Lipp
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Nick Giannoukakis
- Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, Pennsylvania 15212, United States.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Kerry M Empey
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| |
Collapse
|
15
|
Saklani R, Yadav PK, Nengroo MA, Gawali SL, Hassan PA, Datta D, Mishra DP, Dierking I, Chourasia MK. An Injectable In Situ Depot-Forming Lipidic Lyotropic Liquid Crystal System for Localized Intratumoral Drug Delivery. Mol Pharm 2022; 19:831-842. [PMID: 35191706 DOI: 10.1021/acs.molpharmaceut.1c00782] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To address the need for localized chemotherapy against unresectable solid tumors, an injectable in situ depot-forming lipidic lyotropic liquid crystal system (L3CS) is explored that can provide spatiotemporal control over drug delivery. Although liquid crystals have been studied extensively before but their application as an injectable intratumoral depot system for locoregional chemotherapy has not been explored yet. The developed L3CS in the present study is a low-viscosity injectable fluid having a lamellar phase, which transforms into a hexagonal mesophase depot system on subcutaneous or intratumoral injection. The transformed depot system can be preprogrammed to provide tailored drug release intratumorally, over a period of one week to one month. To establish the efficacy of the developed L3CS, doxorubicin is used as a model drug. The drug release mechanism is studied in detail both in vitro and in vivo, and the efficacy of the developed system is investigated in the murine 4T1 tumor model. The direct intratumoral injection of the L3CS provided localized delivery of doxorubicin inside the tumor and restricted its access within the tumor only for a sustained period of time. This led to an over 10-fold reduction in tumor burden, reduced cardiotoxicity, and a significant increase in the median survival rate, compared to the control group. The developed L3CS thus provides an efficient strategy for localized chemotherapy against unresectable solid tumors with a great degree of spatial and temporal control over drug delivery.
Collapse
Affiliation(s)
- Ravi Saklani
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mushtaq A Nengroo
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Santosh L Gawali
- Nanotherapeutics and Biosensors Section, Chemistry Division, Bhabha Atomic Research Centre Trombay, Mumbai 400085, India
| | - Puthusserickal A Hassan
- Nanotherapeutics and Biosensors Section, Chemistry Division, Bhabha Atomic Research Centre Trombay, Mumbai 400085, India
| | - Dipak Datta
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Durga P Mishra
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ingo Dierking
- Department of Physics and Astronomy, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
16
|
Chlorin e6-Biotin Conjugates for Tumor-Targeting Photodynamic Therapy. Molecules 2021; 26:molecules26237342. [PMID: 34885922 PMCID: PMC8658943 DOI: 10.3390/molecules26237342] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
To improve the tumor-targeting efficacy of photodynamic therapy, biotin was conjugated with chlorin e6 to develop a new tumor-targeting photosensitizer, Ce6-biotin. The Ce6-biotin had good water solubility and low aggregation. The singlet-oxygen generation rate of Ce6-biotin was slightly increased compared to Ce6. Flow cytometry and confocal laser scanning microscopy results confirmed Ce6-biotin had higher binding affinity toward biotin-receptor-positive HeLa human cervical carcinoma cells than its precursor, Ce6. Due to the BR-targeting ability of Ce6-biotin, it exhibited stronger cytotoxicity to HeLa cells upon laser irradiation. The IC50 against HeLa cells of Ce6-biotin and Ce6 were 1.28 µM and 2.31 µM, respectively. Furthermore, both Ce6-biotin and Ce6 showed minimal dark toxicity. The selectively enhanced therapeutic efficacy and low dark toxicity suggest that Ce6-biotin is a promising PS for BR-positive-tumor-targeting photodynamic therapy.
Collapse
|
17
|
Taiariol L, Chaix C, Farre C, Moreau E. Click and Bioorthogonal Chemistry: The Future of Active Targeting of Nanoparticles for Nanomedicines? Chem Rev 2021; 122:340-384. [PMID: 34705429 DOI: 10.1021/acs.chemrev.1c00484] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the years, click and bioorthogonal reactions have been the subject of considerable research efforts. These high-performance chemical reactions have been developed to meet requirements not often provided by the chemical reactions commonly used today in the biological environment, such as selectivity, rapid reaction rate, and biocompatibility. Click and bioorthogonal reactions have been attracting increasing attention in the biomedical field for the engineering of nanomedicines. In this review, we study a compilation of articles from 2014 to the present, using the terms "click chemistry and nanoparticles (NPs)" to highlight the application of this type of chemistry for applications involving NPs intended for biomedical applications. This study identifies the main strategies offered by click and bioorthogonal chemistry, with respect to passive and active targeting, for NP functionalization with specific and multiple properties for imaging and cancer therapy. In the final part, a novel and promising approach for "two step" targeting of NPs, called pretargeting (PT), is also discussed; the principle of this strategy as well as all the studies listed from 2014 to the present are presented in more detail.
Collapse
Affiliation(s)
- Ludivine Taiariol
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005 Clermont-Ferrand, France.,Inserm U 1240, F-63000 Clermont-Ferrand, France.,Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| | - Carole Chaix
- Interfaces and Biosensors, UMR 5280, CNRS, F-69100 Villeurbanne, France.,Université de Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Carole Farre
- Interfaces and Biosensors, UMR 5280, CNRS, F-69100 Villeurbanne, France.,Université de Lyon, CNRS, Université Claude Bernard Lyon 1, Institut des Sciences Analytiques, UMR 5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Emmanuel Moreau
- Université Clermont Auvergne, Imagerie Moléculaire et Stratégies Théranostiques, BP 184, F-63005 Clermont-Ferrand, France.,Inserm U 1240, F-63000 Clermont-Ferrand, France.,Centre Jean Perrin, F-63011 Clermont-Ferrand, France
| |
Collapse
|
18
|
Lutein-Loaded, Biotin-Decorated Polymeric Nanoparticles Enhance Lutein Uptake in Retinal Cells. Pharmaceutics 2020; 12:pharmaceutics12090798. [PMID: 32847030 PMCID: PMC7558721 DOI: 10.3390/pharmaceutics12090798] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 01/08/2023] Open
Abstract
Age related macular degeneration (AMD) is one of the leading causes of visual loss and is responsible for approximately 9% of global blindness. It is a progressive eye disorder seen in elderly people (>65 years) mainly affecting the macula. Lutein, a carotenoid, is an antioxidant, and has shown neuroprotective properties in the retina. However, lutein has poor bioavailability owing to poor aqueous solubility. Drug delivery to the posterior segment of the eye is challenging due to the blood–retina barrier. Retinal pigment epithelium (RPE) expresses the sodium-dependent multivitamin transporter (SMVT) transport system which selectively uptakes biotin by active transport. In this study, we aimed to enhance lutein uptake into retinal cells using PLGA–PEG–biotin nanoparticles. Lutein loaded polymeric nanoparticles were prepared using O/W solvent-evaporation method. Particle size and zeta potential (ZP) were determined using Malvern Zetasizer. Other characterizations included differential scanning calorimetry, FTIR, and in-vitro release studies. In-vitro uptake and cytotoxicity studies were conducted in ARPE-19 cells using flow cytometry and confocal microscopy. Lutein was successfully encapsulated into PLGA and PLGA–PEG–biotin nanoparticles (<250 nm) with uniform size distribution and high ZP. The entrapment efficiency of lutein was ≈56% and ≈75% for lutein-loaded PLGA and PLGA–PEG–biotin nanoparticles, respectively. FTIR and DSC confirmed encapsulation of lutein into nanoparticles. Cellular uptake studies in ARPE-19 cells confirmed a higher uptake of lutein with PLGA–PEG–biotin nanoparticles compared to PLGA nanoparticles and lutein alone. In vitro cytotoxicity results confirmed that the nanoparticles were safe, effective, and non-toxic. Findings from this study suggest that lutein-loaded PLGA–PEG–biotin nanoparticles can be potentially used for treatment of AMD for higher lutein uptake.
Collapse
|
19
|
Iannazzo D, Celesti C, Espro C. Recent Advances on Graphene Quantum Dots as Multifunctional Nanoplatforms for Cancer Treatment. Biotechnol J 2020; 16:e1900422. [PMID: 32618417 DOI: 10.1002/biot.201900422] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/11/2020] [Indexed: 12/24/2022]
Abstract
Graphene quantum dots (GQDs), the latest member of the graphene family, have attracted enormous interest in the last few years, due to their exceptional physical, chemical, electrical, optical, and biological properties. Their strong size-dependent photoluminescence and the presence of many reactive groups on the graphene surface allow their multimodal conjugation with therapeutic agents, targeting ligands, polymers, light responsive agents, fluorescent dyes, and functional nanoparticles, making them valuable agents for cancer diagnosis and treatment. In this review, the very recent advances covering the last 3 years on the applications of GQDs as drug delivery systems and theranostic tools for anticancer therapy are discussed, highlighting the relevant factors which regulate their biocompatibility. Among these factors, the size, kind, and degree of surface functionalization have shown to greatly affect their use in biological systems. Toxicity issues, which still represent an open challenge for the clinical development of GQDs based therapeutic agents, are also discussed at cellular and animal levels.
Collapse
Affiliation(s)
- Daniela Iannazzo
- Department of Engineering, University of Messina, Contrada Di Dio, Messina, 98166, Italy
| | - Consuelo Celesti
- Department of Engineering, University of Messina, Contrada Di Dio, Messina, 98166, Italy
| | - Claudia Espro
- Department of Engineering, University of Messina, Contrada Di Dio, Messina, 98166, Italy
| |
Collapse
|
20
|
Wang W, Kong Y, Jiang J, Tian X, Li S, Akshath US, Tiede C, Hondow N, Yu A, Guo Y, Zhou D. Photon induced quantum yield regeneration of cap-exchanged CdSe/CdS quantum rods for ratiometric biosensing and cellular imaging. NANOSCALE 2020; 12:8647-8655. [PMID: 32147673 DOI: 10.1039/c9nr08060k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Full water-dispersion of commercial hydrophobic CdSe/CdS core/shell quantum rods (QRs) was achieved by cap-exchange using a dihydrolipoic acid zwitterion ligand at a low ligand:QR molar ratio (LQMR) of 1000. However, this process almost completely quenched the QR fluorescence, greatly limiting its potential in downstream fluorescence based applications. Fortunately, we found that the QR fluorescence could be recovered by exposure to near ultra-violet to blue light radiation (e.g. 300-450 nm). These "reborn" QRs were found to be compact, bright, and stable, and were resistant to non-specific adsorption, which make them powerful fluorescent probes in broad biomedical applications. We demonstrated their potential in two model applications: first, the QRs were conjugated with His8-tagged small antibody mimetic proteins (also known as Affimers) for the sensitive detection of target proteins via a Förster resonance energy transfer (FRET) readout strategy and second, the QR surface was functionalized with biotins for targeted imaging of cancer cells.
Collapse
Affiliation(s)
- Weili Wang
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Alqaraghuli HGJ, Kashanian S, Rafipour R. A Review on Targeting Nanoparticles for Breast Cancer. Curr Pharm Biotechnol 2020; 20:1087-1107. [PMID: 31364513 DOI: 10.2174/1389201020666190731130001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/11/2022]
Abstract
Chemotherapeutic agents have been used extensively in breast cancer remedy. However, most anticancer drugs cannot differentiate between cancer cells and normal cells, leading to toxic side effects. Also, the resulted drug resistance during chemotherapy reduces treatment efficacy. The development of targeted drug delivery offers great promise in breast cancer treatment both in clinical applications and in pharmaceutical research. Conjugation of nanocarriers with targeting ligands is an effective therapeutic strategy to treat cancer diseases. In this review, we focus on active targeting methods for breast cancer cells through the use of chemical ligands such as antibodies, peptides, aptamers, vitamins, hormones, and carbohydrates. Also, this review covers all information related to these targeting ligands, such as their subtypes, advantages, disadvantages, chemical modification methods with nanoparticles and recent published studies (from 2015 to present). We have discussed 28 different targeting methods utilized for targeted drug delivery to breast cancer cells with different nanocarriers delivering anticancer drugs to the tumors. These different targeting methods give researchers in the field of drug delivery all the information and techniques they need to develop modern drug delivery systems.
Collapse
Affiliation(s)
- Hasanain Gomhor J Alqaraghuli
- Faculty of Chemistry, Razi University, Kermanshah, Iran.,Department of Sciences, College of Basic Education, Al- Muthanna University, Al-Muthanna, Iraq
| | - Soheila Kashanian
- Faculty of Chemistry, Sensor and Biosensor Research Center (SBRC) & Nanoscience and Nanotechnology Research Center (NNRC), Razi University, Kermanshah, Iran.,Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ronak Rafipour
- Department of Chemistry, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| |
Collapse
|
22
|
Oliveira GDFS, Gouveia FS, Pinheiro ADA, do Nascimento Neto LG, de Vasconcelos MA, Teixeira EH, Gondim ACS, Lopes LGDF, de Carvalho IMM, Sousa EHS. An anthracene-pendant ruthenium( ii) complex conjugated to a biotin anchor, an essential handle for photo-induced anti-cancer activity. NEW J CHEM 2020. [DOI: 10.1039/d0nj00209g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Efficient avidin binding and selective cancer cell response upon light irradiation of an enhanced ROS photogenerator biotinylated ruthenium complex.
Collapse
Affiliation(s)
| | - Florencio Sousa Gouveia
- Group of Bioinorganic
- Department of Organic and Inorganic Chemistry
- Federal University of Ceará
- Fortaleza
- Brazil
| | - Aryane de Azevedo Pinheiro
- Laboratório Integrado de Biomoléculas
- Departamento de Patologia e Medicina Legal
- Universidade Federal do Ceará
- Fortaleza
- Brazil
| | | | - Mayron Alves de Vasconcelos
- Laboratório Integrado de Biomoléculas
- Departamento de Patologia e Medicina Legal
- Universidade Federal do Ceará
- Fortaleza
- Brazil
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas
- Departamento de Patologia e Medicina Legal
- Universidade Federal do Ceará
- Fortaleza
- Brazil
| | - Ana Claudia Silva Gondim
- Group of Bioinorganic
- Department of Organic and Inorganic Chemistry
- Federal University of Ceará
- Fortaleza
- Brazil
| | | | | | | |
Collapse
|
23
|
Xu S, Jin Z, Zhang Z, Huang W, Shen Y, Wang Z, Guo S. Precise ratiometric co-loading, co-delivery and intracellular co-release of paclitaxel and curcumin by aid of their conjugation to the same gold nanorods to exert synergistic effects on MCF-7/ADR cells. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
24
|
Biancalana L, Gruchała M, Batchelor LK, Błauż A, Monti A, Pampaloni G, Rychlik B, Dyson PJ, Marchetti F. Conjugating Biotin to Ruthenium(II) Arene Units via Phosphine Ligand Functionalization. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201900922] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Lorenzo Biancalana
- Dipartimento di Chimica e Chimica Industriale Università di Pisa Via G. Moruzzi 13 56124 Pisa Italy
| | - Martyna Gruchała
- Cytometry Lab Department of Molecular Biophysics University of Łódź ul. Pomorska 141/143 90‐236 Łódź Poland
| | - Lucinda K. Batchelor
- Institut des Sciences et Ingénierie Chimiques Ecole Polytechnique Fédérale de Lausanne (EPFL) 1015 Lausanne Switzerland
| | - Andrzej Błauż
- Cytometry Lab Department of Molecular Biophysics University of Łódź ul. Pomorska 141/143 90‐236 Łódź Poland
| | - Andrea Monti
- Dipartimento di Chimica e Chimica Industriale Università di Pisa Via G. Moruzzi 13 56124 Pisa Italy
| | - Guido Pampaloni
- Dipartimento di Chimica e Chimica Industriale Università di Pisa Via G. Moruzzi 13 56124 Pisa Italy
| | - Błażej Rychlik
- Cytometry Lab Department of Molecular Biophysics University of Łódź ul. Pomorska 141/143 90‐236 Łódź Poland
| | - Paul J. Dyson
- Institut des Sciences et Ingénierie Chimiques Ecole Polytechnique Fédérale de Lausanne (EPFL) 1015 Lausanne Switzerland
| | - Fabio Marchetti
- Dipartimento di Chimica e Chimica Industriale Università di Pisa Via G. Moruzzi 13 56124 Pisa Italy
| |
Collapse
|
25
|
Côrte-Real L, Karas B, Brás AR, Pilon A, Avecilla F, Marques F, Preto A, Buckley BT, Cooper KR, Doherty C, Garcia MH, Valente A. Ruthenium-Cyclopentadienyl Bipyridine-Biotin Based Compounds: Synthesis and Biological Effect. Inorg Chem 2019; 58:9135-9149. [PMID: 31241925 DOI: 10.1021/acs.inorgchem.9b00735] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prospective anticancer metallodrugs should consider target-specific components in their design in order to overcome the limitations of the current chemotherapeutics. The inclusion of vitamins, which receptors are overexpressed in many cancer cell lines, has proven to be a valid strategy. Therefore, in this paper we report the synthesis and characterization of a set of new compounds [Ru(η5-C5H5)(P(C6H4R)3)(4,4'-R'-2,2'-bpy)]+ (R = F and R' = H, 3; R = F and R' = biotin, 4; R = OCH3 and R' = H, 5; R = OCH3 and R' = biotin, 6), inspired by the exceptional good results recently obtained for the analogue bearing a triphenylphosphane ligand. The precursors for these syntheses were also described following modified literature procedures, [Ru(η5-C5H5)(P(C6H4R)3)2Cl], where R is -F (1) or -OCH3 (2). The structure of all compounds is fully supported by spectroscopic and analytical techniques and by X-ray diffraction studies for compounds 2, 3, and 5. All cationic compounds are cytotoxic in the two breast cancer cell lines tested, MCF7 and MDA-MB-231, and much better than cisplatin under the same experimental conditions. The cytotoxicity of the biotinylated compounds seems to be related with the Ru uptake by the cells expressing biotin receptors, indicating a potential mediated uptake. Indeed, a biotin-avidin study confirmed that the attachment of biotin to the organometallic fragment still allows biotin recognition by the protein. Therefore, the biotinylated compounds might be potent anticancer drugs as they show cytotoxic effect in breast cancer cells at low dose dependent on the compounds' uptake, induce cell death by apoptosis and inhibit the colony formation of cancer cells causing also less severe side effects in zebrafish.
Collapse
Affiliation(s)
- Leonor Côrte-Real
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Brittany Karas
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States.,Department of Biochemistry and Microbiology , Rutgers University , 76 Lipman Drive , New Brunswick New Jersey 08854 , United States
| | - Ana Rita Brás
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal.,Centre of Molecular and Environmental Biology (CBMA), Department of Biology , University of Minho , Portugal. Campus de Gualtar , Braga 4710-057 , Portugal
| | - Adhan Pilon
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Fernando Avecilla
- Grupo Xenomar, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química, Facultade de Ciencias , Universidade da Coruña , Campus de A Coruña , 15071 A Coruña , Spain
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Te'cnico (C2TN/IST) , Universidade de Lisboa , Estrada Nacional 10 (km 139.7) , 2695-066 Bobadela LRS , Portugal
| | - Ana Preto
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology , University of Minho , Portugal. Campus de Gualtar , Braga 4710-057 , Portugal
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States
| | - Keith R Cooper
- Department of Biochemistry and Microbiology , Rutgers University , 76 Lipman Drive , New Brunswick New Jersey 08854 , United States
| | - Cathleen Doherty
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States
| | - M Helena Garcia
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Andreia Valente
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| |
Collapse
|
26
|
Recent Progress in the Development of Poly(lactic- co-glycolic acid)-Based Nanostructures for Cancer Imaging and Therapy. Pharmaceutics 2019; 11:pharmaceutics11060280. [PMID: 31197096 PMCID: PMC6630460 DOI: 10.3390/pharmaceutics11060280] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
Diverse nanosystems for use in cancer imaging and therapy have been designed and their clinical applications have been assessed. Among a variety of materials available to fabricate nanosystems, poly(lactic-co-glycolic acid) (PLGA) has been widely used due to its biocompatibility and biodegradability. In order to provide tumor-targeting and diagnostic properties, PLGA or PLGA nanoparticles (NPs) can be modified with other functional materials. Hydrophobic or hydrophilic therapeutic cargos can be placed in the internal space or adsorbed onto the surface of PLGA NPs. Protocols for the fabrication of PLGA-based NPs for cancer imaging and therapy are already well established. Moreover, the biocompatibility and biodegradability of PLGA may elevate its feasibility for clinical application in injection formulations. Size-controlled NP’s properties and ligand–receptor interactions may provide passive and active tumor-targeting abilities, respectively, after intravenous administration. Additionally, the introduction of several imaging modalities to PLGA-based NPs can enable drug delivery guided by in vivo imaging. Versatile platform technology of PLGA-based NPs can be applied to the delivery of small chemicals, peptides, proteins, and nucleic acids for use in cancer therapy. This review describes recent findings and insights into the development of tumor-targeted PLGA-based NPs for use of cancer imaging and therapy.
Collapse
|
27
|
Zhang Y, Zhang L, Yin G, Ma W, Li J, Zhou Z, Gao F. In Vivo Pharmacokinetics Assessment of Indocyanine Green-Loaded Nanoparticles in Tumor Tissue with a Dynamic Diffuse Fluorescence Tomography System. Mol Imaging Biol 2019; 21:1044-1053. [PMID: 30850969 DOI: 10.1007/s11307-019-01340-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE The purpose of this study was to show a systematic strategy for assessing the pharmacokinetics of indocyanine green (ICG)-loaded nanoparticles in the tumor tissue based on a dynamic diffuse fluorescence tomography (DFT) system. PROCEDURES Twelve-seven-week-old male Balb/c nude mice bearing HepG2/ADR hepatocellular carcinoma were randomly divided into four groups (n = 3 per group). Four hundred microliters of three types of ICG-loaded nanoparticles (content of ICG: 50 μg/ml) and free ICG (50 μg/ml) was intravenously injected into the mice in each group, respectively. Afterwards, the real-time tomographic images on the spatial level were acquired at 2-11 min, 30 min, 1, 2, 3, 4, 6, 8, 10, 12, and 24 h post-injection, and pharmacokinetic rates were derived for semi-quantitative assessment of the pharmacokinetics of nanoparticles at the tumor site using our proposed pharmacokinetic analysis method. RESULTS The results obtained from our proposed dynamic DFT experiment demonstrated the distribution of different ICG formulations on the spatial level and enabled the semi-quantitative analysis of the pharmacokinetics of nanoparticles in the tumor tissue. CONCLUSIONS The obtained pharmacokinetic rates effectively reflected the metabolic processes of nanoparticles in the tumor tissue, which proves to be beneficial for the development of tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Yanqi Zhang
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China
| | - Limin Zhang
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China. .,Tianjin Key Laboratory of Biomedical Detecting Techniques and Instruments, Tianjin, 300072, China.
| | - Guoyan Yin
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China
| | - Wenjuan Ma
- Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Jiao Li
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Biomedical Detecting Techniques and Instruments, Tianjin, 300072, China
| | - Zhongxing Zhou
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Biomedical Detecting Techniques and Instruments, Tianjin, 300072, China
| | - Feng Gao
- School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, China. .,Tianjin Key Laboratory of Biomedical Detecting Techniques and Instruments, Tianjin, 300072, China.
| |
Collapse
|
28
|
Dual-Targeting Nanoparticles: Codelivery of Curcumin and 5-Fluorouracil for Synergistic Treatment of Hepatocarcinoma. J Pharm Sci 2019; 108:1284-1295. [DOI: 10.1016/j.xphs.2018.10.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/02/2018] [Accepted: 10/16/2018] [Indexed: 12/30/2022]
|
29
|
Kutova OM, Guryev EL, Sokolova EA, Alzeibak R, Balalaeva IV. Targeted Delivery to Tumors: Multidirectional Strategies to Improve Treatment Efficiency. Cancers (Basel) 2019; 11:E68. [PMID: 30634580 PMCID: PMC6356537 DOI: 10.3390/cancers11010068] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
Malignant tumors are characterized by structural and molecular peculiarities providing a possibility to directionally deliver antitumor drugs with minimal impact on healthy tissues and reduced side effects. Newly formed blood vessels in malignant lesions exhibit chaotic growth, disordered structure, irregular shape and diameter, protrusions, and blind ends, resulting in immature vasculature; the newly formed lymphatic vessels also have aberrant structure. Structural features of the tumor vasculature determine relatively easy penetration of large molecules as well as nanometer-sized particles through a blood⁻tissue barrier and their accumulation in a tumor tissue. Also, malignant cells have altered molecular profile due to significant changes in tumor cell metabolism at every level from the genome to metabolome. Recently, the tumor interaction with cells of immune system becomes the focus of particular attention, that among others findings resulted in extensive study of cells with preferential tropism to tumor. In this review we summarize the information on the diversity of currently existing approaches to targeted drug delivery to tumor, including (i) passive targeting based on the specific features of tumor vasculature, (ii) active targeting which implies a specific binding of the antitumor agent with its molecular target, and (iii) cell-mediated tumor targeting.
Collapse
Affiliation(s)
- Olga M Kutova
- The Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., Nizhny Novgorod 603950, Russia.
| | - Evgenii L Guryev
- The Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., Nizhny Novgorod 603950, Russia.
| | - Evgeniya A Sokolova
- The Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., Nizhny Novgorod 603950, Russia.
| | - Razan Alzeibak
- The Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., Nizhny Novgorod 603950, Russia.
| | - Irina V Balalaeva
- The Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., Nizhny Novgorod 603950, Russia.
- The Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya str., Moscow 119991, Russia.
| |
Collapse
|
30
|
Li L, Song Y, He J, Zhang M, Liu J, Ni P. Zwitterionic shielded polymeric prodrug with folate-targeting and pH responsiveness for drug delivery. J Mater Chem B 2019; 7:786-795. [PMID: 32254853 DOI: 10.1039/c8tb02772b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Zwitterionic polymers are a class of polymers that acts as both Lewis base and Lewis acid in solution. These polymers not only have excellent properties of hydration, anti-bacterial adhesion, charge reversal and easy chemical modification, but also have characteristics of long-term circulation and suppress nonspecific protein adsorption in vivo. Here, we describe a novel folate-targeted and acid-labile polymeric prodrug under the microenvironment of tumor cells, abbreviated as FA-P(MPC-co-PEGMA-BZ)-g-DOX, which was synthesized via a combination of reversible addition-fragmentation chain transfer (RAFT) copolymerization, Schiff-base reaction, Click chemistry, and a reaction between the amine group of doxorubicin (DOX) and aldehyde functionalities of P(MPC-co-PEGMA-BZ) pendants, wherein MPC and PEGMA-BZ represent 2-(methacryloyloxy)ethyl phosphorylcholine and polyethylene glycol methacrylate ester benzaldehyde, respectively. The polymeric prodrug could self-assemble into nanoparticles in an aqueous solution. The average particle size and morphologies of the prodrug nanoparticles were observed by dynamic light scattering (DLS) and transmission electron microscopy (TEM), respectively. We also investigated the in vitro drug release behavior and observed rapid prodrug nanoparticle dissociation and drug release under a mildly acidic microenvironment. The methyl thiazolyl tetrazolium (MTT) assay verified that the P(MPC-co-PEGMA-BZ) copolymer possessed good biocompatibility and the FA-P(MPC-co-PEGMA-BZ)-g-DOX prodrug nanoparticles showed higher cellular uptake than those prodrug nanoparticles without the FA moiety. The results of cytotoxicity and the intracellular uptake of non-folate/folate targeted prodrug nanoparticles further confirmed that FA-P(MPC-co-PEGMA-BZ)-g-DOX could be efficiently accumulated and rapidly internalized by HeLa cells due to the strong interaction between multivalent phosphorylcholine (PC) groups and cell membranes. This kind of multifunctional FA-P(MPC-co-PEGMA-BZ)-g-DOX prodrug nanoparticle with combined target-ability and pH responsiveness demonstrates promising potential for cancer chemotherapy.
Collapse
Affiliation(s)
- Lei Li
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou 215123, P. R. China.
| | | | | | | | | | | |
Collapse
|
31
|
Luo J, Meng X, Su J, Ma H, Wang W, Fang L, Zheng H, Qin Y, Chen T. Biotin-Modified Polylactic- co-Glycolic Acid Nanoparticles with Improved Antiproliferative Activity of 15,16-Dihydrotanshinone I in Human Cervical Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9219-9230. [PMID: 30102527 DOI: 10.1021/acs.jafc.8b02698] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
15,16-Dihydrotanshinone I (DI), a natural compound isolated from a traditional Asian functional food Salvia Miltiorrhiza Bunge, is known for its anticancer activity. However, poor solubility of DI limits its desirable anticancer application. Herein, polylactic- co-glycolic acid (PLGA) was functionalized with polyethylene glycol (PEG) and biotin to form copolymers PEG-PLGA (PPA) and biotin-PEG-PLGA (BPA). DI was encapsulated in copolymers PPA and BPA to obtain DI-PPA-NPs (NPs = nanoparticles) and DI-BPA-NPs, respectively. The particle size and its distribution, encapsulation efficiency, and in vitro releasing capacity of DI-BPA-NPs were characterized by biophysical methods. MTT assay was used to evaluate the antiproliferative activity of free DI, DI-PPA-NPs, and DI-BPA-NPs in human cervical cancer Hela cells. DI-BPA-NPs showed the highest cytotoxicity on Hela cells with an IC50 value of 4.55 ± 0.631 μM, while it was 8.20 ± 0.849 and 6.14 ± 0.312 μM for DI and DI-PPA-NPs in 72 h, respectively. The superior antiproliferative activity was supported by the fact that DI-BPA-NPs could be preferentially internalized by Hela cells, owing to their specific interaction between biotin and overexpressed biotin receptors. In addition, DI-BPA-NPs effectively inhibited Hela cell proliferation by inducing G2/M phase cycle arrest and decreasing the intracellular reactive oxygen species (ROS) level by 31.50 ± 2.29% in 5 min. In summary, DI-BPA-NPs shows improved antiproliferative activity against human cervical cancer as comparing with free DI, demonstrating its application potential in cancer therapy.
Collapse
Affiliation(s)
- Jingjing Luo
- School of Food Science and Engineering , South China University of Technology , Guangzhou 510640 , China
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety , Guangzhou 510640 , China
| | - Xiaofeng Meng
- School of Food Science and Engineering , South China University of Technology , Guangzhou 510640 , China
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety , Guangzhou 510640 , China
| | - Jianyu Su
- School of Food Science and Engineering , South China University of Technology , Guangzhou 510640 , China
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety , Guangzhou 510640 , China
- Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center) , Guangzhou 510640 , China
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy , University of Rhode Island , Kingston , Rhode Island 02881 , United States
| | - Wen Wang
- School of Food Science and Engineering , South China University of Technology , Guangzhou 510640 , China
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety , Guangzhou 510640 , China
| | - Liming Fang
- Department of Materials Science and Engineering , South China University of Technology , Guangzhou 510640 , China
| | - Huade Zheng
- Department of Materials Science and Engineering , South China University of Technology , Guangzhou 510640 , China
| | - Yexia Qin
- Institute of Industrial Technology Research , South China University of Technology , Guangzhou 510640 , China
| | - Tianfeng Chen
- Department of Chemistry , Jinan University , Guangzhou , 510632 , China
| |
Collapse
|
32
|
Deshpande NU, Jayakannan M. Biotin-Tagged Polysaccharide Vesicular Nanocarriers for Receptor-Mediated Anticancer Drug Delivery in Cancer Cells. Biomacromolecules 2018; 19:3572-3585. [PMID: 29906389 DOI: 10.1021/acs.biomac.8b00833] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biotin-conjugated multistimuli-responsive polysaccharide vesicular nanocarriers are designed and developed, for the first time, to accomplish receptor-mediated endocytosis in cancer cells and to deliver anticancer drugs to intracellular compartments. For this purpose, a new renewable hydrophobic unit was custom designed with redox-degradable disulfide and enzyme-biodegradable aliphatic ester chemical linkages, and it was conjugated along with biotin on the dextran backbone. The dextran derivative self-assembled into nanovesicles of <200 nm in size, which were characterized by dynamic and static light scattering, electron, and atomic force microscopes. Avidin-HABA assay established the high affinity of biotin-tagged dextran vesicles toward membrane-receptors up to 25 nM concentration. Doxorubicin-hydrochloride (DOX.HCl)-loaded dextran vesicles exhibited stable formulation in phosphate-buffered saline (PBS) and fetal bovine serum (FBS). Redox-degradation by glutathione (GSH) showed 60% drug release, whereas lysosomal esterase enzyme enabled >98% drug release in 12 h. Confocal microscope and flow cytometry-assisted time-dependent cellular uptake studies revealed that the biotin-receptors overexpressed in cervical cancer cells (HeLa) exhibited larger drug accumulation through the receptor-assisted endocytosis process. This process enabled the delivery of higher amount of DOX and significantly enhanced the killing in cancer cells (HeLa) compared to wild-type mouse embryonic fibroblast cells (WT-MEF, normal cells). Control experiments such as biotin pretreatment in cancer cells and energy-suppressed cellular uptake at 4 °C further supported the occurrence of receptor-mediated endocytosis by the biotin-tagged polymer vesicles. This report provides first insights into the targeted polysaccharide vesicle platform, and the proof-of-concept is successfully demonstrated in biotin receptor-overexpressed cervical cancer cells.
Collapse
Affiliation(s)
- Nilesh Umakant Deshpande
- Department of Chemistry , Indian Institute of Science Education and Research (IISER) Pune , Dr. Homi Bhabha Road , Pune 411008 , Maharashtra , India
| | - Manickam Jayakannan
- Department of Chemistry , Indian Institute of Science Education and Research (IISER) Pune , Dr. Homi Bhabha Road , Pune 411008 , Maharashtra , India
| |
Collapse
|
33
|
Lee Y, Lee S, Jon S. Biotinylated Bilirubin Nanoparticles as a Tumor Microenvironment-Responsive Drug Delivery System for Targeted Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800017. [PMID: 29938184 PMCID: PMC6010876 DOI: 10.1002/advs.201800017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/07/2018] [Indexed: 05/03/2023]
Abstract
The tumor microenvironment (TME) plays a crucial role in tumorigenesis and cancer cell metastasis. Accordingly, a drug-delivery system (DDS) that is capable of targeting tumor and releasing drugs in response to TME-associated stimuli should lead to potent antitumor efficacy. Here, a cancer targeting, reactive oxygen species (ROS)-responsive drug delivery vehicle as an example of a TME-targeting DDS is reported. Tumor targeting is achieved using biotin as a ligand for "biotin transporter"-overexpressing malignant tumors, and bilirubin-based nanoparticles (BRNPs) are used as a drug-delivery carrier that enables ROS-responsive drug release. Doxorubicin-loaded, biotinylated BRNPs (Dox@bt-BRNPs) with size of ≈100 nm are prepared by a one-step self-assembly process. Dox@bt-BRNPs exhibit accelerated Dox-release behavior in response to ROS and show specific binding as well as anticancer activity against biotin transporter-overexpressing HeLa cells in vitro. bt-BRNPs labeled with cypate, near-infrared dye, show much greater accumulation at tumor sites in HeLa tumor-bearing mice than BRNPs lacking the biotin ligand. Finally, intravenous injection of Dox@bt-BRNPs into HeLa tumor-bearing mice results in greater antitumor efficacy compared with free Dox, bt-BRNPs only, and Dox@BRNPs without causing any appreciable body weight loss. Collectively, these findings suggest that bt-BRNPs hold potential as a new TME-responsive DDS for effectively treating various tumors.
Collapse
Affiliation(s)
- Yonghyun Lee
- KAIST Institute for the BioCenturyDepartment of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejeon34141Republic of Korea
| | - Soyoung Lee
- KAIST Institute for the BioCenturyDepartment of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejeon34141Republic of Korea
| | - Sangyong Jon
- KAIST Institute for the BioCenturyDepartment of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejeon34141Republic of Korea
| |
Collapse
|
34
|
Colin C, Meyer M, Cerella C, Kleinclauss A, Monard G, Boisbrun M, Diederich M, Flament S, Grillier-Vuissoz I, Kuntz S. Biotinylation enhances the anticancer effects of 15d‑PGJ2 against breast cancer cells. Int J Oncol 2018; 52:1991-2000. [PMID: 29620161 DOI: 10.3892/ijo.2018.4338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/08/2018] [Indexed: 11/05/2022] Open
Abstract
15-Deoxy-∆12,14-prostaglandin J2 (15d‑PGJ2) is a natural agonist of peroxisome proliferator-activated receptor γ (PPARγ) that displays anticancer activity. Various studies have indicated that the effects of 15d‑PGJ2 are due to both PPARγ-dependent and -independent mechanisms. In the present study, we examined the effects of a biotinylated form of 15d‑PGJ2 (b‑15d‑PGJ2) on hormone-dependent MCF‑7 and triple‑negative MDA‑MB‑231 breast cancer cell lines. b‑15d‑PGJ2 inhibited cell proliferation more efficiently than 15d‑PGJ2 or the synthetic PPARγ agonist, efatutazone. b‑15d‑PGJ2 was also more potent than its non-biotinylated counterpart in inducing apoptosis. We then analyzed the mechanisms underlying this improved efficiency. It was found not to be the result of biotin receptor-mediated increased incorporation, since free biotin in the culture medium did not decrease the anti-proliferative activity of b‑15d‑PGJ2 in competition assays. Of note, b‑15d‑PGJ2 displayed an improved PPARγ agonist activity, as measured by transactivation experiments. Molecular docking analyses revealed a similar insertion of b‑15d‑PGJ2 and 15d‑PGJ2 into the ligand binding domain of PPARγ via a covalent bond with Cys285. Finally, PPARγ silencing markedly decreased the cleavage of the apoptotic markers, poly(ADP-ribose) polymerase 1 (PARP‑1) and caspase‑7, that usually occurs following b‑15d‑PGJ2 treatment. Taken together, our data indicate that biotinylation enhances the anti-proliferative and pro-apoptotic activity of 15d‑PGJ2, and that this effect is partly mediated via a PPARγ-dependent pathway. These results may aid in the development of novel therapeutic strategies for breast cancer treatment.
Collapse
Affiliation(s)
| | - Maxime Meyer
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France
| | - Claudia Cerella
- Laboratory for Molecular and Cellular Biology of Cancer, Kirchberg Hospital, L‑2540 Luxembourg, Luxembourg
| | | | - Gérald Monard
- Université de Lorraine, CNRS, LPCT, F-54000 Nancy, France
| | | | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151‑742, Republic of Korea
| | | | | | - Sandra Kuntz
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France
| |
Collapse
|
35
|
Liu X, Gong P, Song P, Xie F, Miller Ii AL, Chen S, Lu L. Fast functionalization of ultrasound microbubbles using strain promoted click chemistry. Biomater Sci 2018; 6:623-632. [PMID: 29411006 PMCID: PMC5829049 DOI: 10.1039/c8bm00004b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Functionalization of microbubbles (MBs) is a difficult issue due to their unstable nature. Here we report a fast and versatile method using a strain promoted alkyne-azide cycloaddition (SPAAC) click reaction for microbubble functionalization. An azadibenzocyclooctyne (DBCO) group was first introduced onto the MB surface and then an azide group into the desired ligand. Without any initiators or catalysts, essential click ligation occurred within 1 min and a majority of the reaction completed in 5 min at 37 °C. This fast ligation shortens the microbubble reaction time and preserves essential amounts of microbubbles for further in situ imaging and delivery of therapeutics.
Collapse
Affiliation(s)
- Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Biotin conjugated organic molecules and proteins for cancer therapy: A review. Eur J Med Chem 2018; 145:206-223. [PMID: 29324341 DOI: 10.1016/j.ejmech.2018.01.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Accepted: 01/01/2018] [Indexed: 01/09/2023]
Abstract
The main transporter for biotin is sodium dependent multivitamin transporter (SMVT), which is overexpressed in various aggressive cancer cell lines such as ovarian (OV 2008, ID8), leukemia (L1210FR), mastocytoma (P815), colon (Colo-26), breast (4T1, JC, MMT06056), renal (RENCA, RD0995), and lung (M109) cancer cell lines. Furthermore, its overexpression was found higher to that of folate receptor. Therefore, biotin demand in the rapidly growing tumors is higher than normal tissues. Several biotin conjugated organic molecules has been reported here for selective delivery of the drug in cancer cell. Biotin conjugated molecules are showing higher fold of cytotoxicity in biotin positive cancer cell lines than the normal cell. Nanoparticles and polymer surface modified drugs and biotin mediated cancer theranostic strategy was highlighted in this review. The cytotoxicity and selectivity of the drug in cancer cells has enhanced after biotin conjugation.
Collapse
|