1
|
Rathee A, Solanki P, Emad NA, Zai I, Ahmad S, Alam S, Alqahtani AS, Noman OM, Kohli K, Sultana Y. Posaconazole-hemp seed oil loaded nanomicelles for invasive fungal disease. Sci Rep 2024; 14:16588. [PMID: 39025925 PMCID: PMC11258229 DOI: 10.1038/s41598-024-66074-1] [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: 04/09/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Invasive fungal infections (IFI) pose a significant health burden, leading to high morbidity, mortality, and treatment costs. This study aims to develop and characterize nanomicelles for the codelivery of posaconazole and hemp seed oil for IFI via the oral route. The nanomicelles were prepared using a nanoprecipitation method and optimized through the Box Behnken design. The optimized nanomicelles resulted in satisfactory results for zeta potential, size, PDI, entrapment efficiency, TEM, and stability studies. FTIR and DSC results confirm the compatibility and amorphous state of the prepared nanomicelles. Confocal laser scanning microscopy showed that the optimized nanomicelles penetrated the tissue more deeply (44.9µm) than the suspension (25µm). The drug-loaded nanomicelles exhibited sustained cumulative drug release of 95.48 ± 3.27% for 24 h. The nanomicelles showed significant inhibition against Aspergillus niger and Candida albicans (22.4 ± 0.21 and 32.2 ± 0.46 mm, respectively). The pharmacokinetic study on Wistar rats exhibited a 1.8-fold increase in relative bioavailability for the nanomicelles compared to the suspension. These results confirm their therapeutic efficacy and lay the groundwork for future research and clinical applications, providing a promising synergistic antifungal nanomicelles approach for treating IFIs.
Collapse
Affiliation(s)
- Anjali Rathee
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Pavitra Solanki
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Nasr A Emad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Iqra Zai
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Saeem Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Shadab Alam
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Ali S Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, 11451, Riyadh, Saudi Arabia
| | - Omar M Noman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, 11451, Riyadh, Saudi Arabia
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India
| | - Yasmin Sultana
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, 110062, New Delhi, India.
| |
Collapse
|
2
|
Yazdan M, Naghib SM, Mozafari MR. Polymeric Micelle-Based Nanogels as Emerging Drug Delivery Systems in Breast Cancer Treatment: Promises and Challenges. Curr Drug Targets 2024; 25:649-669. [PMID: 38919076 DOI: 10.2174/0113894501294136240610061328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024]
Abstract
Breast cancer is a pervasive global health issue that disproportionately impacts the female population. Over the past few years, there has been considerable interest in nanotechnology due to its potential utility in creating drug-delivery systems designed to combat this illness. The primary aim of these devices is to enhance the delivery of targeted medications, optimise the specific cells that receive the drugs, tackle treatment resistance in malignant cells, and introduce novel strategies for preventing and controlling diseases. This research aims to examine the methodologies utilised by various carrier nanoparticles in the context of therapeutic interventions for breast cancer. The main objective is to investigate the potential application of novel delivery technologies to attain timely and efficient diagnosis and treatment. Current cancer research predominantly examines diverse drug delivery methodologies for chemotherapeutic agents. These methodologies encompass the development of hydrogels, micelles, exosomes, and similar compounds. This research aims to analyse the attributes, intricacies, notable advancements, and practical applications of the system in clinical settings. Despite the demonstrated efficacy of these methodologies, an apparent discrepancy can be observed between the progress made in developing innovative therapeutic approaches and their widespread implementation in clinical settings. It is critical to establish a robust correlation between these two variables to enhance the effectiveness of medication delivery systems based on nanotechnology in the context of breast cancer treatment.
Collapse
Affiliation(s)
- M Yazdan
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - S M Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
3
|
Chandra J, Hasan N, Nasir N, Wahab S, Thanikachalam PV, Sahebkar A, Ahmad FJ, Kesharwani P. Nanotechnology-empowered strategies in treatment of skin cancer. ENVIRONMENTAL RESEARCH 2023; 235:116649. [PMID: 37451568 DOI: 10.1016/j.envres.2023.116649] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
In current scenario skin cancer is a serious condition that has a significant impact on world health. Skin cancer is divided into two categories: melanoma skin cancer (MSC) and non-melanoma skin cancer (NMSC). Because of its significant psychosocial effects and need for significant investment in new technology and therapies, skin cancer is an illness of global health relevance. From the patient's perspective chemotherapy considered to be the most acceptable form of treatment. However, significant negatives of chemotherapy such as severe toxicities and drug resistance pose serious challenges to the treatment. The field of nanomedicine holds significant promise for enhancing the specificity of targeting neoplastic cells through the facilitation of targeted drug delivery to tumour cells. The integration of multiple therapeutic modalities to selectively address cancer-promoting or cell-maintaining pathways constitutes a fundamental aspect of cancer treatment. The use of mono-therapy remains prevalent in the treatment of various types of cancer, it is widely acknowledged in the academic community that this conventional approach is generally considered to be less efficacious compared to the combination treatment strategy. The employment of combination therapy in cancer treatment has become increasingly widespread due to its ability to produce synergistic anticancer effects, mitigate toxicity associated with drugs, and inhibit multi-drug resistance by means of diverse mechanisms. Nanotechnology based combination therapy represents a promising avenue for the development of efficacious therapies for skin cancer within the context of this endeavour. The objective of this article is to provide a description of distinct challenges for efficient delivery of drugs via skin. This article also provides a summary of the various nanotechnology based combinatorial therapy available for skin cancer with their recent advances. This review also focuses on current status of clinical trials of such therapies.
Collapse
Affiliation(s)
- Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazim Nasir
- Department of Basic Medical Sciences, College of Applied Medical Sciences, Khamis Mushait, Kingdom of Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 61421, Saudi Arabia
| | - Punniyakoti Veeraveedu Thanikachalam
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
4
|
Rathee A, Solanki P, Verma S, Vohora D, Ansari MJ, Aodah A, Kohli K, Sultana Y. Simultaneous Determination of Posaconazole and Hemp Seed Oil in Nanomicelles through RP-HPLC via a Quality-by-Design Approach. ACS OMEGA 2023; 8:30057-30067. [PMID: 37636934 PMCID: PMC10448652 DOI: 10.1021/acsomega.3c02097] [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: 03/29/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023]
Abstract
The present study involves the development of a reverse-phase HPLC method employing the quality-by-design methodology for the estimation of posaconazole and hemp seed oil simultaneously in nanomicelles formulation. The successful separation of posaconazole and hemp seed oil was achieved together, and this is the first study to develop and quantify posaconazole and hemp seed oil nanomicelles with linoleic acid as the internal standard and developed a dual drug analytical method employing a quality-by-design approach. The study was performed on a Shimadzu Prominence-I LC-2030C 3D Plus HPLC system with a PDA detector and the Shim-pack Solar C8 column (250 mm × 4.6 mm × 5 μm) for analysis with a mobile phase ratio of methanol:water (80:20% v/v) maintaining the flow rate of 1.0 mL/min. The final wavelength was selected as 240 nm and the elution of hemp seed oil and posaconazole was obtained at 2.7 and 4.6 min, respectively, with a maximum run time of 8.0 min. Box Behnken design was employed to optimize the method, keeping the retention time, peak area, and theoretical plates as dependent variables, while the mobile phase composition, flow rate, and wavelengths were chosen as independent variables. Parameters such as specificity, accuracy, robustness, linearity, sensitivity, precision, ruggedness, and forced degradation study were performed to validate the method. The calibration curves of posaconazole and hemp seed oil were determined to be linear throughout the range for concentration. The suggested approach can be effectively utilized for estimating the content of drugs from their nanoformulation and proved suitable for both in vivo and in vitro research.
Collapse
Affiliation(s)
- Anjali Rathee
- Department
of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Pavitra Solanki
- Department
of Pharmaceutics, Delhi Pharmaceutical Sciences
and Research University, Pushp Vihar, Sec-III, New Delhi 110017, India
| | - Surajpal Verma
- Department
of Pharmaceutical Analysis, Delhi Pharmaceutical
Sciences and Research University, Pushp Vihar, Sec-III, New Delhi 110017, India
| | - Divya Vohora
- Department
of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Javed Ansari
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdul Aziz University, Al-kharj 11231, Saudi Arabia
| | - Alhussain Aodah
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdul Aziz University, Al-kharj 11231, Saudi Arabia
| | - Kanchan Kohli
- Lloyd Institute
of Management and Technology, Greater Noida, Uttar Pradesh 201306, India
| | - Yasmin Sultana
- Department
of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
5
|
Cheng T, Tai Z, Shen M, Li Y, Yu J, Wang J, Zhu Q, Chen Z. Advance and Challenges in the Treatment of Skin Diseases with the Transdermal Drug Delivery System. Pharmaceutics 2023; 15:2165. [PMID: 37631379 PMCID: PMC10458513 DOI: 10.3390/pharmaceutics15082165] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Skin diseases are among the most prevalent non-fatal conditions worldwide. The transdermal drug delivery system (TDDS) has emerged as a promising approach for treating skin diseases, owing to its numerous advantages such as high bioavailability, low systemic toxicity, and improved patient compliance. However, the effectiveness of the TDDS is hindered by several factors, including the barrier properties of the stratum corneum, the nature of the drug and carrier, and delivery conditions. In this paper, we provide an overview of the development of the TDDS from first-generation to fourth-generation systems, highlighting the characteristics of each carrier in terms of mechanism composition, penetration method, mechanism of action, and recent preclinical studies. We further investigated the significant challenges encountered in the development of the TDDS and the crucial significance of clinical trials.
Collapse
Affiliation(s)
- Tingting Cheng
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (T.C.); (J.Y.); (J.W.)
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Min Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Ying Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Junxia Yu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (T.C.); (J.Y.); (J.W.)
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Jiandong Wang
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (T.C.); (J.Y.); (J.W.)
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| | - Zhongjian Chen
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (T.C.); (J.Y.); (J.W.)
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; (Z.T.); (M.S.); (Y.L.)
| |
Collapse
|
6
|
Liu L, Zhao W, Ma Q, Gao Y, Wang W, Zhang X, Dong Y, Zhang T, Liang Y, Han S, Cao J, Wang X, Sun W, Ma H, Sun Y. Functional nano-systems for transdermal drug delivery and skin therapy. NANOSCALE ADVANCES 2023; 5:1527-1558. [PMID: 36926556 PMCID: PMC10012846 DOI: 10.1039/d2na00530a] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/27/2022] [Indexed: 06/18/2023]
Abstract
Transdermal drug delivery is one of the least intrusive and patient-friendly ways for therapeutic agent administration. Recently, functional nano-systems have been demonstrated as one of the most promising strategies to treat skin diseases by improving drug penetration across the skin barrier and achieving therapeutically effective drug concentrations in the target cutaneous tissues. Here, a brief review of functional nano-systems for promoting transdermal drug delivery is presented. The fundamentals of transdermal delivery, including skin biology and penetration routes, are introduced. The characteristics of functional nano-systems for facilitating transdermal drug delivery are elucidated. Moreover, the fabrication of various types of functional transdermal nano-systems is systematically presented. Multiple techniques for evaluating the transdermal capacities of nano-systems are illustrated. Finally, the advances in the applications of functional transdermal nano-systems for treating different skin diseases are summarized.
Collapse
Affiliation(s)
- Lijun Liu
- School of Pharmacy, Qingdao University Qingdao 266071 China
- The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University Qingdao 266021 China
| | - Wenbin Zhao
- School of Pharmacy, Qingdao University Qingdao 266071 China
- The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University Qingdao 266021 China
| | - Qingming Ma
- School of Pharmacy, Qingdao University Qingdao 266071 China
- The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University Qingdao 266021 China
| | - Yang Gao
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Weijiang Wang
- School of Pharmacy, Qingdao University Qingdao 266071 China
- The Shandong Consortium in the Yellow River Basin for Prevention, Treatment and Drug Development for Primary Diseases Related to Alcoholism, Qingdao University Qingdao 266021 China
| | - Xuan Zhang
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Yunxia Dong
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Tingting Zhang
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Yan Liang
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Shangcong Han
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Jie Cao
- School of Pharmacy, Qingdao University Qingdao 266071 China
| | - Xinyu Wang
- Institute of Thermal Science and Technology, Shandong University Jinan 250061 China
| | - Wentao Sun
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences Qingdao 266113 China
| | - Haifeng Ma
- Department of Geriatrics, Zibo Municipal Hospital Zibo 255400 China
| | - Yong Sun
- School of Pharmacy, Qingdao University Qingdao 266071 China
| |
Collapse
|
7
|
Prajapat VM, Mahajan S, Paul PG, Aalhate M, Mehandole A, Madan J, Dua K, Chellappan DK, Singh SK, Singh PK. Nanomedicine: A pragmatic approach for tackling melanoma skin cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
8
|
Al-Obaidy R, Haider AJ, Al-Musawi S, Arsad N. Targeted delivery of paclitaxel drug using polymer-coated magnetic nanoparticles for fibrosarcoma therapy: in vitro and in vivo studies. Sci Rep 2023; 13:3180. [PMID: 36823237 PMCID: PMC9950487 DOI: 10.1038/s41598-023-30221-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Fibrosarcoma is a rare type of cancer that affects cells known as fibroblasts that are malignant, locally recurring, and spreading tumor in fibrous tissue. In this work, an iron plate immersed in an aqueous solution of double added deionized water, supplemented with potassium permanganate solution (KMnO4) was carried out by the pulsed laser ablation in liquid method (PLAIL). Superparamagnetic iron oxide nanoparticles (SPIONs) were synthesized using different laser wavelengths (1064, 532, and 266 nm) at a fluence of 28 J/cm2 with 100 shots of the iron plate to control the concentration, shape and size of the prepared high-stability SPIONs. The drug nanocarrier was synthesized by coating SPION with paclitaxel (PTX)-loaded chitosan (Cs) and polyethylene glycol (PEG). This nanosystem was functionalized by receptors that target folate (FA). The physiochemical characteristics of SPION@Cs-PTX-PEG-FA nanoparticles were evaluated and confirmed by Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM), transmission electron microscopy (TEM), X-Ray diffraction (XRD), atomic force microscopy (AFM), and dynamic light scattering (DLS) methods. Cell internalization, cytotoxicity assay (MTT), apoptosis induction, and gene expression of SPION@Cs-PTX-PEG-FA were estimated in fibrosarcoma cell lines, respectively. In vivo studies used BALB/c tumor-bearing mice. The results showed that SPION@Cs-PTX-PEG-FA exhibited suitable physical stability, spherical shape, desirable size, and charge. SPION@Cs-PTX-PEG-FA inhibited proliferation and induced apoptosis of cancer cells (P < 0.01). The results of the in vivo study showed that SPION@Cs-PTX-PEG-FA significantly decreased tumor size compared to free PTX and control samples (P < 0.05), leading to longer survival, significantly increased splenocyte proliferation and IFN-γ level, and significantly decreased the level of IL-4. All of these findings indicated the potential of SPION@Cs-PTX-PEG-FA as an antitumor therapeutic agent.
Collapse
Affiliation(s)
- Rusul Al-Obaidy
- grid.444967.c0000 0004 0618 8761Applied Sciences Department/Laser Science and Technology Branch, University of Technology, Baghdad, Iraq
| | - Adawiya J. Haider
- grid.444967.c0000 0004 0618 8761Applied Sciences Department/Laser Science and Technology Branch, University of Technology, Baghdad, Iraq
| | | | - Norhana Arsad
- Photonics Technology Laboratory, Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia UKM, 43600, Bangi, Malaysia.
| |
Collapse
|
9
|
Mukerabigwi JF, Tang R, Cao Y, Mohammed F, Zhou Q, Zhou M, Ge Z. Mitochondria-Targeting Polyprodrugs to Overcome the Drug Resistance of Cancer Cells by Self-Amplified Oxidation-Triggered Drug Release. Bioconjug Chem 2023; 34:377-391. [PMID: 36716444 DOI: 10.1021/acs.bioconjchem.2c00559] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The multi-drug resistance (MDR) of cancers is one of the main barriers for the success of diverse chemotherapeutic methods and is responsible for most cancer deaths. Developing efficient approaches to overcome MDR is still highly desirable for efficient chemotherapy of cancers. The delivery of targeted anticancer drugs that can interact with mitochondrial DNA is recognized as an effective strategy to reverse the MDR of cancers due to the relatively weak DNA-repairing capability in the mitochondria. Herein, we report on a polyprodrug that can sequentially target cancer cells and mitochondria using folic acid (FA) and tetraphenylphosphonium (TPP) targeting moieties, respectively. They were conjugated to the terminal groups of the amphiphilic block copolymer prodrugs composed of poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) and copolymerized monomers containing cinnamaldehyde (CNM) and doxorubicin (DOX). After self-assembly into micelles with the suitable size (∼30 nm), which were termed as TF@CNM + DOX, and upon intravenous administration, the micelles can accumulate in tumor tissues. After FA-mediated endocytosis, the endosomal acidity (∼pH 5) can trigger the release of CNM from TF@CNM + DOX micelles, followed by enhanced accumulation into the mitochondria via the TPP target. This promotes the overproduction of reactive oxygen species (ROS), which can subsequently enhance the intracellular oxidative stress and trigger ROS-responsive release of DOX into the mitochondria. TF@CNM + DOX shows great potential to inhibit the growth of DOX-resistant MCF-7 ADR tumors without observable side effects. Therefore, the tumor and mitochondria dual-targeting polyprodrug design represents an ideal strategy to treat MDR tumors through improvement of the intracellular oxidative level and ROS-responsive drug release.
Collapse
Affiliation(s)
- Jean Felix Mukerabigwi
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.,Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Kigali, 3900 Kigali, Rwanda
| | - Rui Tang
- Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yufei Cao
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.,CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Min Zhou
- Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
10
|
Verma P, Gupta GD, Markandeywar TS, Singh D. A Critical Sojourn of Polymeric Micelles: Technological Concepts, Recent Advances, and Future Prospects. Assay Drug Dev Technol 2023; 21:31-47. [PMID: 36856457 DOI: 10.1089/adt.2022.079] [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: 03/02/2023] Open
Abstract
Poorly soluble drug molecules/phytoconstituents are still a growing concern for biopharmaceutical delivery in the body. Polymeric micelles are the amphiphilic block copolymers and have been widely investigated as targeted nanocarriers for the treatment of various ailments. The versatility of nanocarriers is the self-assembling properties in the aqueous medium and forms a stable isotropic system in vivo. The hydrophobic core-hydrophilic shell configuration of the polymers used to the mixed micelles makes easy encapsulation of hydrophobic and hydrophilic drugs into the core. Polymeric micelles can also be combined with targeting ligands that increase their uptake by specific cells, decreasing off-target effects, and provide enhanced therapeutic effect. In the present review, we primarily focused on a critical appraisal of Polymeric micelles along with the method of preparation, mechanism of micelle formulation, and the ongoing formulations under clinical trials. In addition, the biological applications of this isotropic nanocarrier have been duly presented in each route of administration along with suitable case studies.
Collapse
Affiliation(s)
- Princi Verma
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
11
|
Wu Y, Zhang Z, Wei Y, Qian Z, Wei X. Nanovaccines for cancer immunotherapy: Current knowledge and future perspectives. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
12
|
Recent developments of nanomedicine delivery systems for the treatment of pancreatic cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Chaudhuri A, Ramesh K, Kumar DN, Dehari D, Singh S, Kumar D, Agrawal AK. Polymeric micelles: A novel drug delivery system for the treatment of breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
14
|
Farjadian F, Ghasemi S, Akbarian M, Hoseini-Ghahfarokhi M, Moghoofei M, Doroudian M. Physically stimulus-responsive nanoparticles for therapy and diagnosis. Front Chem 2022; 10:952675. [PMID: 36186605 PMCID: PMC9515617 DOI: 10.3389/fchem.2022.952675] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Nanoparticles offer numerous advantages in various fields of science, particularly in medicine. Over recent years, the use of nanoparticles in disease diagnosis and treatments has increased dramatically by the development of stimuli-responsive nano-systems, which can respond to internal or external stimuli. In the last 10 years, many preclinical studies were performed on physically triggered nano-systems to develop and optimize stable, precise, and selective therapeutic or diagnostic agents. In this regard, the systems must meet the requirements of efficacy, toxicity, pharmacokinetics, and safety before clinical investigation. Several undesired aspects need to be addressed to successfully translate these physical stimuli-responsive nano-systems, as biomaterials, into clinical practice. These have to be commonly taken into account when developing physically triggered systems; thus, also applicable for nano-systems based on nanomaterials. This review focuses on physically triggered nano-systems (PTNSs), with diagnostic or therapeutic and theranostic applications. Several types of physically triggered nano-systems based on polymeric micelles and hydrogels, mesoporous silica, and magnets are reviewed and discussed in various aspects.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| | - Soheila Ghasemi
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| | - Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | | | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| |
Collapse
|
15
|
Gao F, Chen Z, Zhou L, Xiao X, Wang L, Liu X, Wang C, Guo Q. Preparation, characterization and in vitro study of bellidifolin nano-micelles. RSC Adv 2022; 12:21982-21989. [PMID: 36043071 PMCID: PMC9364364 DOI: 10.1039/d2ra02779h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/09/2022] [Indexed: 11/21/2022] Open
Abstract
Bellidifolin (BEL), a xanthone compound, has significant therapeutic effectiveness in cardiac diseases such as arrhythmias. However, BEL is limited in clinical applications by its hydrophobicity. In this work, we used BEL as the active pharmaceutical ingredient (API), and polyethylene glycol 15-hydroxy stearate (Kolliphor HS15) as the carrier to prepare BEL nano-micelles by a solvent-volatilization method. According to an analysis by differential scanning calorimetry (DSC), BEL was successfully encapsulated in HS15 as BEL nano-micelles with a 90% encapsulation rate, and particle size was 12.60 ± 0.074 nm in the shape of a sphere and electric potential was −4.76 ± 4.47 mV with good stability and sustained release characteristics. In addition, compared with free drugs, these nano-micelles can increase cellular uptake capacity, inhibit the proliferation of human cardiac fibroblasts, and down-regulate the expression of Smad-2, α-SMA, Collagen I, and Collagen III proteins in myocardial cells to improve myocardial fibrosis. In conclusion, the BEL nano-micelles can provide a new way for the theoretical basis for the clinical application of anti-cardiac fibrosis. Bellidifolin (BEL), a xanthone compound, has significant therapeutic effectiveness in cardiac diseases such as arrhythmias.![]()
Collapse
Affiliation(s)
- Fan Gao
- Hebei TCM Formula Preparation Technology Innovation Center, Hebei University of Chinese Medicine Shijiazhuang 050091 People's Republic of China
| | - Ziyue Chen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine Tianjin 301617 People's Republic of China
| | - Li Zhou
- Hebei TCM Formula Preparation Technology Innovation Center, Hebei University of Chinese Medicine Shijiazhuang 050091 People's Republic of China
| | - Xuefeng Xiao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine Tianjin 301617 People's Republic of China
| | - Lin Wang
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research Tianjin 300301 People's Republic of China
| | - Xingchao Liu
- Hebei TCM Formula Preparation Technology Innovation Center, Hebei University of Chinese Medicine Shijiazhuang 050091 People's Republic of China
| | - Chenggang Wang
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research Tianjin 300301 People's Republic of China
| | - Qiuhong Guo
- Hebei TCM Formula Preparation Technology Innovation Center, Hebei University of Chinese Medicine Shijiazhuang 050091 People's Republic of China
| |
Collapse
|
16
|
Chaturvedi S, Garg A. A comprehensive review on novel delivery approaches for exemestane. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Nguyen NT, Bui QA, Huynh PD, Nguyen QH, Tran NQ, Viet NT, Nguyen DT. Curcumin and Paclitaxel co-Loaded Heparin and Poloxamer P403 Hybrid Nanocarrier for Improved Synergistic Efficacy in Breast Cancer. Curr Drug Deliv 2022; 19:966-979. [DOI: 10.2174/1567201819666220401095923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/22/2022]
Abstract
Introduction:
Multi-drug nanosystem has been employed in several therapeutic models due to the synergistic effect of the drugs and/or bioactive compounds, which help in tumor-targeting and limit usual side effects of chemotherapy.
Methods:
In this research, we developed the amphiphilic Heparin-Poloxamer P403 (HSP) nanogel that can load curcumin (CUR) and Paclitaxel (PTX) through the hydrophobic core of Poloxamer P403. The features of HSP nanogel are assessed through Fourier-transform infrared spectroscopy (FT-IR), transmission electron microscopy (TEM), differential light scattering (DLS), and critical micelle concentration (CMC). Nanogel and its duel-loaded platform show high stability and spherical morphology.
Results:
The drug release profile indicates fast release at pH 5.5, suggesting effective drug distribution at the tumor site. In vitro research confirms lower cytotoxicity of HSP@CUR@PTX compared with free PTX and higher inhibition effect with MCF-7 than HSP@PTX. These results support the synergism between PTX and CUR.
Conclusion,:
HSP@CUR@PTX suggests a prominent strategy for achieving the synergistic effect of PTX and CUR to circumvent undesirable effects in breast cancer treatment.
Collapse
Affiliation(s)
- Ngoc The Nguyen
- Faculty of Medicine - Pharmacy, Tra Vinh University, Tra Vinh City, Vietnam
| | - Quynh Anh Bui
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Phuong Duy Huynh
- Faculty of Medicine - Pharmacy, Tra Vinh University, Tra Vinh City, Vietnam
| | | | - Ngoc Quyen Tran
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam;
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi City, Vietnam
| | - Nguyen Thanh Viet
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Dinh Trung Nguyen
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| |
Collapse
|
18
|
Khan NH, Mir M, Qian L, Baloch M, Ali Khan MF, Rehman AU, Ngowi EE, Wu DD, Ji XY. Skin cancer biology and barriers to treatment: Recent applications of polymeric micro/nanostructures. J Adv Res 2022; 36:223-247. [PMID: 35127174 PMCID: PMC8799916 DOI: 10.1016/j.jare.2021.06.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background Skin cancer has been the leading type of cancer worldwide. Melanoma and non-melanoma skin cancers are now the most common types of skin cancer that have been reached to epidemic proportion. Based on the rapid prevalence of skin cancers, and lack of efficient drug delivery systems, it is essential to surge the possible ways to prevent or cure the disease. Aim of review Although surgical modalities and therapies have been made great progress in recent years, however, there is still an urgent need to alleviate its increased burden. Hence, understanding the precise pathophysiological signaling mechanisms and all other factors of such skin insults will be beneficial for the development of more efficient therapies. Key scientific concepts of review In this review, we explained new understandings about onset and development of skin cancer and described its management via polymeric micro/nano carriers-based therapies, highlighting the current key bottlenecks and future prospective in this field. In therapeutic drug/gene delivery approaches, polymeric carriers-based system is the most promising strategy. This review discusses that how polymers have successfully been exploited for development of micro/nanosized systems for efficient delivery of anticancer genes and drugs overcoming all the barriers and limitations associated with available conventional therapies. In addition to drug/gene delivery, intelligent polymeric nanocarriers platforms have also been established for combination anticancer therapies including photodynamic and photothermal, and for theranostic applications. This portfolio of latest approaches could promote the blooming growth of research and their clinical availability.
Collapse
Key Words
- 5-ALA, 5-aminolevulinic acid
- 5-FU, 5-fluorouracil
- AIDS, Acquired immune deficiency syndrome
- BCC, Basal cell carcinoma
- BCCs, Basal cell carcinomas
- Basal cell carcinoma
- CREB, response element-binding protein
- DDS, Drug delivery system
- DIM-D, Di indolyl methane derivative
- Drug delivery
- GNR-PEG-MN, PEGylated gold nanorod microneedle
- Gd, Gadolinium
- Gene delivery
- HH, Hedgehog
- HPMC, Hydroxypropyl methylcellulose
- IPM, Isopropyl myristate
- MCIR, Melanocortin-1 receptor
- MNPs, Magnetic nanoparticle
- MNs, Microneedles
- MRI, Magnetic Resonance Imaging
- MSC, Melanoma skin cancer
- Microneedles
- Mn, Manganese
- NMSC, Non melanoma skin cancer
- NPs, Nano Particles
- OTR, Organ transplant recipients
- PAMAM, Poly-amidoamines
- PAN, Polyacrylonitrile
- PATCH1, Patch
- PCL, Poly (ε-caprolactone)
- PDT, Photodynamic therapy
- PEG, Polyethylene glycol
- PLA, Poly lactic acid
- PLA-HPG, Poly (d-l-lactic acid)-hyperbranched polyglycerol
- PLGA, Poly (lactide-co-glycolide) copolymers
- PLL, Poly (L-lysine)
- Polymeric nanocarriers
- QDs, Quantum dots
- SC, Skin cancer
- SCC, Squamous cell Carcinoma
- SMO, Smoothen
- SPIO, Superparamagnetic iron oxide
- Squamous cell carcinoma
- UV, Ultra Violet
- cAMP, Cyclic adenosine monophosphate
- dPG, Dendritic polyglycerol
- hTERT, Human telomerase reverse transcriptase
Collapse
Affiliation(s)
- Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Life Sciences. Henan University, Kaifeng, Henan 475004, China
| | - Maria Mir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Lei Qian
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Mahnoor Baloch
- School of Natural Sciences, National University of Science and Technology, Islamabad 44000, Pakistan
| | - Muhammad Farhan Ali Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim-ur- Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- Department of Biological Sciences, Faculty of Sciences, Dar es Salaam University College of Education, Dar es Salaam 2329, Tanzania
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| |
Collapse
|
19
|
Nano Drug Delivery Systems: Effective Therapy Strategies to Overcome Multidrug Resistance in Tumor Cells. ChemistrySelect 2022. [DOI: 10.1002/slct.202104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Xian C, Chen H, Xiong F, Fang Y, Huang H, Wu J. Platinum-based chemotherapy via nanocarriers and co-delivery of multiple drugs. Biomater Sci 2021; 9:6023-6036. [PMID: 34323260 DOI: 10.1039/d1bm00879j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Platinum-based anticancer drugs can inhibit the growth of cancer cells by disrupting DNA replication, which makes them widely applicable in clinics for treating tumors and cancers. However, owing to the intrinsic or acquired drug resistance and severe side effects caused in the treatment, their successful clinical applications have been limited. Various strategies have been used to address these challenges. Nanocarriers have been used for platinum drug delivery because they can be effectively deposited in tumor tissues to reduce the damage to normal organs for an enhanced permeability and retention (EPR) effect. Furthermore, for synergizing the function of platinum-based drugs with different mechanisms to decrease the toxicities, multicomponent chemotherapy has become an imperative strategy in clinical cancer treatments. This review aims to introduce the mechanisms of action and limitations of platinum-based drugs in clinics, followed by providing the current advancement of nanocarriers including lipids, polymers, dendrimers, micelles and albumin for platinum drug delivery in cancer treatments. In addition, multicomponent chemotherapy based on platinum drugs is introduced in detail. Finally, the prospects of multicomponent chemotherapy for cancer treatment are discussed as well.
Collapse
Affiliation(s)
- Caihong Xian
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518057, China
| | - Haolin Chen
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518057, China
| | - Fei Xiong
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518057, China
| | - Yifen Fang
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518057, China
| |
Collapse
|
21
|
Zuccari G, Alfei S, Zorzoli A, Marimpietri D, Turrini F, Baldassari S, Marchitto L, Caviglioli G. Increased Water-Solubility and Maintained Antioxidant Power of Resveratrol by Its Encapsulation in Vitamin E TPGS Micelles: A Potential Nutritional Supplement for Chronic Liver Disease. Pharmaceutics 2021; 13:pharmaceutics13081128. [PMID: 34452090 PMCID: PMC8400607 DOI: 10.3390/pharmaceutics13081128] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
Children affected by chronic liver disease exhibit impaired neurocognitive development and growth due to the low absorption and digestion of nutrients. Furthermore, malnutrition is an adverse prognostic factor in liver transplantation as it is associated with an increase in morbidity and mortality. D-α-tocopheryl-polyethylene-glycol-succinate (TPGS) is currently administered per os as a vitamin E source to improve children's survival and well-being; however, TPGS alone does not reverse spinocerebellar degeneration and lipid peroxidation. To potentiate the effects of TPGS, we loaded micelles with resveratrol (RES), a natural polyphenol, with antioxidant and antiinflammatory activities, which has demonstrated protective action in the liver. Firstly, we investigated the suitability of TPGS to encapsulate RES in micelles by means of a phase-solubility study, then RES-TPGS formulations were prepared via solvent casting and solvent diffusion evaporation methods. RES-TPGS colloidal dispersions showed small mean diameters (12 nm), low polydispersity, and quite neutral Zeta potentials. The formulations showed a sustained drug release and a good drug loading capacity, further confirmed by infrared spectroscopy and differential scanning calorimetry. RES-TPGSs exhibited unaltered antioxidant activity compared to pristine RES via the DPPH assay and a significant reduction in toxicity compared to empty TPGS on HaCaT cells. Thus, RES-TPGS micelles may overcome the challenges of current liver disease therapy by providing more protective effects thanks to the antioxidant activity of RES and by reducing the surfactant toxicity on normal cells.
Collapse
Affiliation(s)
- Guendalina Zuccari
- Department of Pharmacy (DiFAR), University of Genoa, Viale Cembrano 4-I, 16148 Genova, Italy; (S.A.); (F.T.); (S.B.); (G.C.)
- Correspondence:
| | - Silvana Alfei
- Department of Pharmacy (DiFAR), University of Genoa, Viale Cembrano 4-I, 16148 Genova, Italy; (S.A.); (F.T.); (S.B.); (G.C.)
| | - Alessia Zorzoli
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (A.Z.); (D.M.)
| | - Danilo Marimpietri
- Stem Cell Laboratory and Cell Therapy Center, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy; (A.Z.); (D.M.)
| | - Federica Turrini
- Department of Pharmacy (DiFAR), University of Genoa, Viale Cembrano 4-I, 16148 Genova, Italy; (S.A.); (F.T.); (S.B.); (G.C.)
| | - Sara Baldassari
- Department of Pharmacy (DiFAR), University of Genoa, Viale Cembrano 4-I, 16148 Genova, Italy; (S.A.); (F.T.); (S.B.); (G.C.)
| | - Leonardo Marchitto
- Department of Sciences for the Quality of Life, University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy;
| | - Gabriele Caviglioli
- Department of Pharmacy (DiFAR), University of Genoa, Viale Cembrano 4-I, 16148 Genova, Italy; (S.A.); (F.T.); (S.B.); (G.C.)
| |
Collapse
|
22
|
da Silva Leite JM, Patriota YBG, de La Roca MF, Soares-Sobrinho JL. New Perspectives in Drug Delivery Systems for the Treatment of Tuberculosis. Curr Med Chem 2021; 29:1936-1958. [PMID: 34212827 DOI: 10.2174/0929867328666210629154908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Tuberculosis is a chronic respiratory disease caused by Mycobacterium tuberculosis. The common treatment regimens of tuberculosis are lengthy with adverse side effects, low patient compliance, and antimicrobial resistance. Drug delivery systems (DDSs) can overcome these limitations. OBJECTIVE This review aims to summarize the latest DDSs for the treatment of tuberculosis. In the first section, the main pharmacokinetic and pharmacodynamic challenges, due to the innate properties of the drugs, are put forth. The second section elaborates on the use of DDS to overcome the disadvantages of the current treatment of tuberculosis. CONCLUSION We reviewed research articles published in the last 10 years. DDSs can improve the physicochemical properties of anti-tuberculosis drugs, improving solubility, stability, and bioavailability, with better control of drug release and can target alveolar macrophages. However, more preclinical studies and robust bio-relevant analyses are needed for DDSs to become a feasible option to treat patients and attract investors.
Collapse
Affiliation(s)
- Joandra Maísa da Silva Leite
- Núcleo de Controle de Qualidade de Medicamentos e Correlatos, Federal University of Pernambuco, Recife, PE, Brazil
| | - Yuri Basilio Gomes Patriota
- Núcleo de Controle de Qualidade de Medicamentos e Correlatos, Federal University of Pernambuco, Recife, PE, Brazil
| | - Mônica Felts de La Roca
- Núcleo de Controle de Qualidade de Medicamentos e Correlatos, Federal University of Pernambuco, Recife, PE, Brazil
| | | |
Collapse
|
23
|
Villamizar-Sarmiento MG, Guerrero J, Moreno-Villoslada I, Oyarzun-Ampuero FA. The key role of the drug self-aggregation ability to obtain optimal nanocarriers based on aromatic-aromatic drug-polymer interactions. Eur J Pharm Biopharm 2021; 166:19-29. [PMID: 34052430 DOI: 10.1016/j.ejpb.2021.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/20/2021] [Accepted: 05/23/2021] [Indexed: 11/18/2022]
Abstract
The efficient association and controlled release of hydrophilic and aromatic low molecular-weight drugs (HALMD) still remains a challenge due to their relatively weak interactions with excipients and strong affinity to water. Considering that a wide variety of drugs to treat chronic diseases are HALMD, their inclusion in polymeric nanoparticles (NPs) constitutes an attractive possibility by providing to these drugs with controllable physiochemical properties, preventing crisis episodes, decreasing dose-dependent side effects and promoting therapeutic adhesiveness. However, the strong interaction of HALMD with the aqueous medium jeopardizes their encapsulation and controlled release. In this work, the role of the self-assembly tendency of HALMD on their association with the aromatic excipient poly(sodium 4-styrensulfonate) (PSS) to form NPs is studied. For this aim, the widely used drugs amitriptyline (AMT), promethazine (PMZ), and chlorpheniramine (CPM) are selected due to their well described critical aggregation concentration (cac) (36 mM for AMT, 36 mM for PMZ, and 69.5 mM for CPM). These drugs undergo aromatic-aromatic interactions with the polymer, which stabilize their mutual binding, as seen by NMR. The simple mixing of solutions of opposite charged molecules (drug + PSS) allowed obtaining NPs. Importantly, comparing the three drugs, the formation of NPs occurred at significantly lower absolute concentration and significantly lower drug/polymer ratio as the cac takes lower values, indicating a stronger binding to the polymer, as also deduced from the respective drug/polymer dissociation constant values. In addition, the number of formed NPs is similar for all formulations, even though a much lower concentration of the drug and polymer is present in systems comprising lower cac. The obtained NPs are spheroidal and present size between 100 and 160 nm, low polydispersity (≤0.3) and negative zeta potential (from -30 to -60 mV). The association efficiency reaches values ≥ 83% and drug loading could achieve values up to 68% (never evidenced before for systems comprising HALMD). In addition, drug release studies are also significantly influenced by cac, providing more prolonged release for AMT and PMZ (lower cac), whose delivery profiles adjust to the Korsmeyer-Peppas equation. As a novelty of this work, a synergic contribution of drug self-association tendency and aromatic-aromatic interaction between the drug and polymers is highlighted, a fact that could be crucial for the rational design and development of efficient drug delivery systems.
Collapse
Affiliation(s)
- María Gabriela Villamizar-Sarmiento
- Department of Sciences and Pharmaceutical Technology, University of Chile, Santiago de Chile 8380494, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile.
| | - Juan Guerrero
- Laboratorio de Compuestos de Coordinación y Química Supramolecular, Facultad de Química y Biología, Universidad de Santiago de Chile, Av. Libertador Bernardo O'Higgins 3363, Estación central, 9170002 Santiago, Chile.
| | - Ignacio Moreno-Villoslada
- Instituto de Ciencias Químicas, Facultad de Ciencias, Universidad Austral de Chile, Casilla 567, Valdivia 5110033, Chile.
| | - Felipe A Oyarzun-Ampuero
- Department of Sciences and Pharmaceutical Technology, University of Chile, Santiago de Chile 8380494, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile; Center of New Drugs for Hypertension (CENDHY), Universidad de Chile & Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Wan Z, Zheng R, Moharil P, Liu Y, Chen J, Sun R, Song X, Ao Q. Polymeric Micelles in Cancer Immunotherapy. Molecules 2021; 26:1220. [PMID: 33668746 PMCID: PMC7956602 DOI: 10.3390/molecules26051220] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapies have generated some miracles in the clinic by orchestrating our immune system to combat cancer cells. However, the safety and efficacy concerns of the systemic delivery of these immunostimulatory agents has limited their application. Nanomedicine-based delivery strategies (e.g., liposomes, polymeric nanoparticles, silico, etc.) play an essential role in improving cancer immunotherapies, either by enhancing the anti-tumor immune response, or reducing their systemic adverse effects. The versatility of working with biocompatible polymers helps these polymeric nanoparticles stand out as a key carrier to improve bioavailability and achieve specific delivery at the site of action. This review provides a summary of the latest advancements in the use of polymeric micelles for cancer immunotherapy, including their application in delivering immunological checkpoint inhibitors, immunostimulatory molecules, engineered T cells, and cancer vaccines.
Collapse
Affiliation(s)
- Zhuoya Wan
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (Z.W.); (J.C.); (X.S.)
| | - Ruohui Zheng
- Department of Pharmaceutical Science, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Pearl Moharil
- Department of Cell Biology, Harvard Medical School, Harvard University, Boston, MA 02115, USA;
| | - Yuzhe Liu
- Department of Materials Engineering, Purdue University, West Lafayette, IN 47906, USA;
| | - Jing Chen
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (Z.W.); (J.C.); (X.S.)
- Department of Pharmaceutical Science, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Runzi Sun
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Xu Song
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (Z.W.); (J.C.); (X.S.)
| | - Qiang Ao
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; (Z.W.); (J.C.); (X.S.)
| |
Collapse
|
25
|
Reduction-triggered di-block copolymer prodrug for high-performance long-acting tumor-selective killing. Colloids Surf B Biointerfaces 2020; 196:111368. [DOI: 10.1016/j.colsurfb.2020.111368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 01/19/2023]
|
26
|
Ramanunny AK, Wadhwa S, Gulati M, Singh SK, Kapoor B, Dureja H, Chellappan DK, Anand K, Dua K, Khursheed R, Awasthi A, Kumar R, Kaur J, Corrie L, Pandey NK. Nanocarriers for treatment of dermatological diseases: Principle, perspective and practices. Eur J Pharmacol 2020; 890:173691. [PMID: 33129787 DOI: 10.1016/j.ejphar.2020.173691] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
Skin diseases are the fourth leading non-fatal skin conditions that act as a burden and affect the world economy globally. This condition affects the quality of a patient's life and has a pronounced impact on both their physical and mental state. Treatment of these skin conditions with conventional approaches shows a lack of efficacy, long treatment duration, recurrence of conditions, systemic side effects, etc., due to improper drug delivery. However, these pitfalls can be overcome with the applications of nanomedicine-based approaches that provide efficient site-specific drug delivery at the target site. These nanomedicine-based strategies are evolved as potential treatment opportunities in the form of nanocarriers such as polymeric and lipidic nanocarriers, nanoemulsions along with emerging others viz. carbon nanotubes for dermatological treatment. The current review focuses on challenges faced by the existing conventional treatments along with the topical therapeutic perspective of nanocarriers in treating various skin diseases. A total of 213 articles have been reviewed and the application of different nanocarriers in treating various skin diseases has been explained in detail through case studies of previously published research works. The toxicity related aspects of nanocarriers are also discussed.
Collapse
Affiliation(s)
| | - Sheetu Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Dinesh Kumar Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Rubiya Khursheed
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Rajan Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Jaskiran Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Narendra Kumar Pandey
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| |
Collapse
|
27
|
Thymoquinone-Loaded Soluplus ®-Solutol ® HS15 Mixed Micelles: Preparation, In Vitro Characterization, and Effect on the SH-SY5Y Cell Migration. Molecules 2020; 25:molecules25204707. [PMID: 33066549 PMCID: PMC7587349 DOI: 10.3390/molecules25204707] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Thymoquinone (TQ) is the main active ingredient of Nigella sativa essential oil, with remarkable anti-neoplastic activities with anti-invasive and anti-migratory abilities on a variety of cancer cell lines. However, its poor water solubility, high instability in aqueous solution and pharmacokinetic drawbacks limits its use in therapy. Soluplus® and Solutol® HS15 were employed as amphiphilic polymers for developing polymeric micelles (SSM). Chemical and physical characterization studies of micelles are reported, in terms of size, homogeneity, zeta potential, critical micelle concentration (CMC), cloud point, encapsulation efficiency (EE%), load capacity (DL), in vitro release, and stability. This study reports for the first time the anti-migratory activity of TQ and TQ loaded in SSM (TQ-SSM) in the SH-SY5Y human neuroblastoma cell line. The inhibitory effect was assessed by the wound-healing assay and compared with that of the unformulated TQ. The optimal TQ-SSM were provided with small size (56.71 ± 1.41 nm) and spherical shape at ratio of 1:4 (Soluplus:Solutol HS15), thus increasing the solubility of about 10-fold in water. The entrapment efficiency and drug loading were 92.4 ± 1.6% and 4.68 ± 0.12, respectively, and the colloidal dispersion are stable during storage for a period of 40 days. The TQ-SSM were also lyophilized to obtain a more workable product and with increased stability. In vitro release study indicated a prolonged release of TQ. In conclusion, the formulation of TQ into SSM allows a bio-enhancement of TQ anti-migration activity, suggesting that TQ-SSM is a better candidate than unformulated TQ to inhibit human SH-SY5Y neuroblastoma cell migration.
Collapse
|
28
|
Liao Z, Wong SW, Yeo HL, Zhao Y. Smart nanocarriers for cancer treatment: Clinical impact and safety. NANOIMPACT 2020; 20:100253. [DOI: 10.1016/j.impact.2020.100253] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
29
|
Yang P, Zhang L, Wang T, Liu Q, Wang J, Wang Y, Tu Z, Lin F. Doxorubicin and Edelfosine Combo-Loaded Lipid-Polymer Hybrid Nanoparticles for Synergistic Anticancer Effect Against Drug-Resistant Osteosarcoma. Onco Targets Ther 2020; 13:8055-8067. [PMID: 32884291 PMCID: PMC7434523 DOI: 10.2147/ott.s259428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/13/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction The failure of chemotherapy in osteosarcoma results in drug resistance and acute side effects in the body. Methods In this study, we have prepared a novel folate receptor-targeted doxorubicin (DOX) and edelfosine (EDL)-loaded lipid-polymer hybrid nanoparticle (DE-FPLN) to enhance the anticancer efficacy in osteosarcoma. The nanoparticles were thoroughly characterized for in vitro biological assays followed by detailed antitumor efficacy analysis and toxicity analysis in a xenograft model. Results The dual drug-loaded nanoparticles showed a nanosized morphology and physiological stability. The targeted nanoparticles showed enhanced cellular internalization and subcellular distribution in MG63 cancer cells compared to that of non-targeted nanoparticles. Among many ratios of DOX and EDL, 1:1 ratiometric combinations of drugs were observed to be highly synergistic in killing the cancer cells. MTT assay and caspase-3/7 activity assay clearly showed the superior anticancer efficacy of DE-FPLN formulations in inducing the cancer cell death. In vitro results indicate that the co-administration of two drugs in a folic acid-targeted nanoparticle could potentially induce the apoptosis and cell death. In vivo results displayed the potency of tumor cell killing and significant suppression of tumor growth without any detectable side effects. Conclusion The lipid-polymer hybrid nanocarriers with multiple properties of high drug loading, sequential and ratiometric drug release, improved physiological stability, prolonged blood circulation, and tumor-specific targeting are promising for the delivery of multiple drugs in the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Ping Yang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Lian Zhang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Tian Wang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Qi Liu
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Jing Wang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Yaling Wang
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Zhiquan Tu
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Feng Lin
- Department of Oncology, The Eighth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| |
Collapse
|
30
|
Li S, Zhao W, Liang N, Xu Y, Kawashima Y, Sun S. Multifunctional micelles self-assembled from hyaluronic acid conjugate for enhancing anti-tumor effect of paclitaxel. REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2020.104608] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Advances in anti-breast cancer drugs and the application of nano-drug delivery systems in breast cancer therapy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101662] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Wu DM, Liu T, Deng SH, Han R, Zhang T, Li J, Xu Y. Alpha-1 Antitrypsin Induces Epithelial-to-Mesenchymal Transition, Endothelial-to-Mesenchymal Transition, and Drug Resistance in Lung Cancer Cells. Onco Targets Ther 2020; 13:3751-3763. [PMID: 32440144 PMCID: PMC7210034 DOI: 10.2147/ott.s242579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose Alpha-1 antitrypsin (A1AT) is a secreted protein that plays an important role in various diseases. However, the role of A1AT in non-small cell lung cancer is obscure. Materials and Methods A1AT expression in non-small cell lung cancer was analyzed using quantitative reverse transcription PCR, Western blotting (WB), immunohistochemistry (IHC), and ELISA. WB and IF were used to analyze markers of epithelial-to-mesenchymal transition (EMT), EndoMT, and cancer stem cell (CSC). Transwell and cell wound healing assays were used to analyze migration and invasion abilities. Colony formation and CCK-8 assays were used to analyze cell proliferation following cisplatin treatment. Results A1AT expression was higher in lung cancer samples than in normal tissues and the increased expression was correlated with poor overall survival of patients. In vitro experiments showed that A1AT overexpressed by plasmid transfection significantly promoted migration, invasion, EMT, EndoMT, stemness, and colony formation in lung cancer cell lines, as opposed to A1AT downregulation by siRNA transfection, which significantly inhibited all these variables. Conclusion A1AT is a novel therapeutic target and might be associated with tumor metastasis in lung carcinoma.
Collapse
Affiliation(s)
- Dong-Ming Wu
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Teng Liu
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Shi-Hua Deng
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Rong Han
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Ting Zhang
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Jing Li
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| | - Ying Xu
- Clinical Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
33
|
Zhao YC, Zheng HL, Wang XR, Zheng XL, Chen Y, Fei WD, Zheng YQ, Wang WX, Zheng CH. Enhanced Percutaneous Delivery of Methotrexate Using Micelles Prepared with Novel Cationic Amphipathic Material. Int J Nanomedicine 2020; 15:3539-3550. [PMID: 32547012 PMCID: PMC7245457 DOI: 10.2147/ijn.s251431] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/05/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Methotrexate (MTX) is an antiproliferative drug widely used to treat inflammatory diseases and autoimmune diseases. The application of percutaneous administration is hindered due to its poor transdermal penetration. To reduce side effects and enhanced percutaneous delivery of MTX, novel methotrexate (MTX)-loaded micelles prepared with a amphiphilic cationic material, N,N-dimethyl-(N',N'-di-stearoyl-1-ethyl)1,3-diaminopropane (DMSAP), was designed. MATERIALS AND METHODS DMSAP was synthesized via three steps using simple chemical agents. H nuclear magnetic resonance and mass spectroscopy were used to confirm the successful synthesis of DMSAP. A safe and non-toxic phosphatidylcholine, soybean phosphatidylcholine (SPC), was added to DMSAP at different ratios to form P/D-micelles. Then, MTX-entrapped micelles (M/P/D-micelles) were prepared by electrostatic adsorption. The physicochemical properties and blood stability of micelles were examined thoroughly. In addition, the transdermal potential of the micelles was evaluated by permeation experiments. RESULTS In aqueous environments, DMSAP conjugates could self-assemble spontaneously into micelles with a low critical micelle concentration (CMC) of 0.056 mg/mL. Stable, spherical MTX-entrapped micelles (M/P/D-micelles) with a size of 100-120 nm and high zeta potential of +36.26 mV were prepared. In vitro permeation studies showed that M/P/D-micelles exhibited superior skin permeability and deposition of MTX in the epidermis and dermis compared with that of free MTX. CONCLUSION These special novel cationic M/P/D-micelles can enhance the permeability of MTX and are expected to be a promising percutaneous delivery system for therapy skin diseases.
Collapse
Affiliation(s)
- Yun-Chun Zhao
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Hai-Li Zheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, People’s Republic of China
| | - Xiao-Rong Wang
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Xiao-Ling Zheng
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Yue Chen
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Wei-Dong Fei
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Yong-Quan Zheng
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| | - Wen-Xi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, People’s Republic of China
| | - Cai-Hong Zheng
- Pharmacy Department, Zhejiang University, Women’s Hospital, School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
34
|
Yang F, Xu J, Fu M, Ji J, Chi L, Zhai G. Development of stimuli-responsive intelligent polymer micelles for the delivery of doxorubicin. J Drug Target 2020; 28:993-1011. [PMID: 32378974 DOI: 10.1080/1061186x.2020.1766474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Doxorubicin is still used as a first-line drug in current therapeutics for numerous types of malignant tumours (including lymphoma, transplantable leukaemia and solid tumour). Nevertheless, to overcome the serious side effects like cardiotoxicity and myelosuppression caused by effective doses of doxorubicin remains as a world-class puzzle. In recent years, the usage of biocompatible polymeric nanomaterials to form an intelligently sensitive carrier for the targeted release in tumour microenvironment has attracted wide attention. These different intelligent polymeric micelles (PMs) could change the pharmacokinetics process of drugs or respond in the special microenvironment of tumour site to maximise the efficacy and reduce the toxicity of doxorubicin in other tissues and organs. Several intelligent PMs have already been in the clinical research stage and planned for market. Therefore, related research remains active, and the latest nanotechnology approaches for doxorubicin delivery are always in the spotlight. Centring on the model drugs doxorubicin, this review summarised the mechanisms of PMs, classified the polymers used in the application of doxorubicin delivery and discussed some interesting and imaginative smart PMs in recent years.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jiangkang Xu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Manfei Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Liqun Chi
- Department of Pharmacy, Haidian Maternal and Child Health Hospital of Beijing, Beijing, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
35
|
Yang XL, Wu WX, Li J, Hu ZE, Wang N, Yu XQ. A facile strategy to construct fluorescent pH-sensitive drug delivery vehicle. POLYMER 2020. [DOI: 10.1016/j.polymer.2020.122496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
36
|
Liu Y, Dai S, Wen L, Zhu Y, Tan Y, Qiu G, Meng T, Yu F, Yuan H, Hu F. Enhancing Drug Delivery for Overcoming Angiogenesis and Improving the Phototherapy Efficacy of Glioblastoma by ICG-Loaded Glycolipid-Like Micelles. Int J Nanomedicine 2020; 15:2717-2732. [PMID: 32368051 PMCID: PMC7184138 DOI: 10.2147/ijn.s234240] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
Background Phototherapy is a potential new candidate for glioblastoma (GBM) treatment. However inadequate phototherapy due to stability of the photosensitizer and low target specificity induces the proliferation of neovascular endothelial cells for angiogenesis and causes poor prognosis. Methods In this study, we constructed c(RGDfk)-modified glycolipid-like micelles (cRGD-CSOSA) encapsulating indocyanine green (ICG) for dual-targeting neovascular endothelial cells and tumor cells, and cRGD-CSOSA/ICG mediated dual effect of PDT/PTT with NIR irradiation. Results In vitro, cRGD-CSOSA/ICG inhibited cell proliferation and blocked angiogenesis with NIR irradiation. In vivo, cRGD-CSOSA/ICG exhibited increased accumulation in neovascular endothelial cells and tumor cells. Compared with that of CSOSA, the accumulation of cRGD-CSOSA in tumor tissue was further improved after dual-targeted phototherapy pretreatment. With NIR irradiation, the tumor-inhibition rate of cRGD-CSOSA/ICG was 80.00%, significantly higher than that of ICG (9.08%) and CSOSA/ICG (42.42%). Histological evaluation showed that the tumor vessels were reduced and that the apoptosis of tumor cells increased in the cRGD-CSOSA/ICG group with NIR irradiation. Conclusion The cRGD-CSOSA/ICG nanoparticle-mediated dual-targeting phototherapy could enhance drug delivery to neovascular endothelial cells and tumor cells for anti-angiogenesis and improve the phototherapy effect of glioblastoma, providing a new strategy for glioblastoma treatment.
Collapse
Affiliation(s)
- Yupeng Liu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Suhuan Dai
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Lijuan Wen
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China.,National Engineering Research Center for Modernization of Traditional Chinese Medicine - Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 342700, People's Republic of China
| | - Yun Zhu
- Ocean College, Zhejiang University, Zhoushan 316021, Republic of China
| | - Yanan Tan
- Ocean College, Zhejiang University, Zhoushan 316021, Republic of China
| | - Guoxi Qiu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Fangying Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| |
Collapse
|
37
|
Preparation of fluorophore-tagged polymeric drug delivery vehicles with multiple biological stimuli-triggered drug release. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 108:110358. [DOI: 10.1016/j.msec.2019.110358] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/07/2019] [Accepted: 10/21/2019] [Indexed: 02/04/2023]
|
38
|
Comparison of Chitosan Nanoparticles and Soluplus Micelles to Optimize the Bioactivity of Posidonia oceanica Extract on Human Neuroblastoma Cell Migration. Pharmaceutics 2019; 11:pharmaceutics11120655. [PMID: 31817615 PMCID: PMC6955792 DOI: 10.3390/pharmaceutics11120655] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Posidonia oceanica (L.) Delile is a marine plant endemic of Mediterranean Sea endowed with interesting bioactivities. The hydroalcholic extract of P. oceanica leaves (POE), rich in polyphenols and carbohydrates, has been shown to inhibit human cancer cell migration. Neuroblastoma is a common childhood extracranial solid tumor with high rate of invasiveness. Novel therapeutics loaded into nanocarriers may be used to target the migratory and metastatic ability of neuroblastoma. Our goal was to improve both the aqueous solubility of POE and its inhibitory effect on cancer cell migration. Methods: Chitosan nanoparticles (NP) and Soluplus polymeric micelles (PM) loaded with POE have been developed. Nanoformulations were chemically and physically defined and characterized. In vitro release studies were also performed. Finally, the inhibitory effect of both nanoformulations was tested on SH-SY5Y cell migration by wound healing assay and compared to that of unformulated POE. Results: Both nanoformulations showed excellent physical and chemical stability during storage, and enhanced the solubility of POE. PM-POE improved the inhibitory effect of POE on cell migration probably due to the high encapsulation efficiency and the prolonged release of the extract. Conclusions: For the first time, a phytocomplex of marine origin, i.e., P. oceanica extract, has enhanced in terms of acqueous solubility and bioactivity once encapsulated inside nanomicelles.
Collapse
|
39
|
Li ZP, Tian GX, Jiang H, Pan RY, Lian B, Wang M, Gao ZQ, Zhang B, Wu JL. Liver-Targeting and pH-Sensitive Sulfated Hyaluronic Acid Mixed Micelles for Hepatoma Therapy. Int J Nanomedicine 2019; 14:9437-9452. [PMID: 31819442 PMCID: PMC6896933 DOI: 10.2147/ijn.s214528] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Background The tumor-targeting ability and pH-sensitive properties of intelligent drug delivery systems are crucial for effective drug delivery and anti-tumor therapy. Methods In this study, sHA-DOX/HA-GA mixed micelles were designed with the following properties: sulfated hyaluronic acid (sHA) was synthesized to block cell migration by inhibiting HAase; sHA-DOX conjugates were synthesized via pH-sensitive hydrazone bond to realize DOX-sensitive release. The introduction of HA-GA conjugate could improve active-targeting ability and cellular uptake. Results The results showed that the mixed micelles possessed a nearly spherical shape, nanoscale particle size (217.70±0.89 nm), narrow size distribution (PDI=0.07±0.04), negative zeta potential (-31.87±0.61 mV) and pH-dependent DOX release. In addition, the sHA-DOX/HA-GA micelles exhibited concentration-dependent cytotoxicities against liver carcinoma cells (HepG2) and HeLa cells, and were shown to be effectively taken up by HepG2 cells by confocal microscopy analysis. Furthermore, the in vivo anti-tumor study showed that mixed micelles had a superior anti-tumor effect compared to that of free DOX. Further evidence obtained from the hematoxylin-eosin staining and immunohistochemistry analysis also demonstrated that sHA-DOX/HA-GA exhibited stronger tumor inhibition and lower systemic toxicity than free DOX. Conclusion The sHA-DOX/HA-GA mixed micelles could be a potential drug delivery system for anti-hepatoma therapy.
Collapse
Affiliation(s)
- Zhi-Peng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Gui-Xiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Hong Jiang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Rui-Yan Pan
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Min Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Zhi-Qin Gao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Jing-Liang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| |
Collapse
|
40
|
Zhao MD, Li JQ, Chen FY, Dong W, Wen LJ, Fei WD, Zhang X, Yang PL, Zhang XM, Zheng CH. Co-Delivery of Curcumin and Paclitaxel by "Core-Shell" Targeting Amphiphilic Copolymer to Reverse Resistance in the Treatment of Ovarian Cancer. Int J Nanomedicine 2019; 14:9453-9467. [PMID: 31819443 PMCID: PMC6898996 DOI: 10.2147/ijn.s224579] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/07/2019] [Indexed: 12/27/2022] Open
Abstract
Background Ovarian cancer is a common malignancy in the female reproductive system with a high mortality rate. The most important reason is multidrug resistance (MDR) of cancer chemotherapy. To reduce side effects, reverse resistance and improve efficacy for the treatment of ovarian cancer, a “core-shell” polymeric nanoparticle-mediated curcumin and paclitaxel co-delivery platform was designed. Methods Nuclear magnetic resonance confirmed the successful grafting of polyethylenimine (PEI) and stearic acid (SA) (PEI-SA), which is designed as a mother core for transport carrier. Then, PEI-SA was modified with hyaluronic acid (HA) and physicochemical properties were examined. To understand the regulatory mechanism of resistance and measure the anti-tumor efficacy of the treatments, cytotoxicity assay, cellular uptake, P-glycoprotein (P-gp) expression and migration experiment of ovarian cancer cells were performed. In addition, adverse reactions of nanoformulation to the reproductive system were examined. Results HA-modified drug-loaded PEI-SA had a narrow size of about 189 nm in diameters, and the particle size was suitable for endocytosis. The nanocarrier could target specifically to CD44 receptor on the ovarian cancer cell membrane. Co-delivery of curcumin and paclitaxel by the nanocarriers exerts synergistic anti-ovarian cancer effects on chemosensitive human ovarian cancer cells (SKOV3) and multi-drug resistant variant (SKOV3-TR30) in vitro, and it also shows a good anti-tumor effect in ovarian tumor-bearing nude mice. The mechanism of reversing drug resistance may be that the nanoparticles inhibit the efflux of P-gp, inhibit the migration of tumor cells, and curcumin synergistically reverses the resistance of PTX to increase antitumor activity. It is worth noting that the treatment did not cause significant toxicity to the uterus and ovaries with the observation of macroscopic and microscopic. Conclusion This special structure of targeting nanoparticles co-delivery with the curcumin and paclitaxel can increase the anti-tumor efficacy without increasing the adverse reactions as a promising strategy for therapy ovarian cancer.
Collapse
Affiliation(s)
- Meng-Dan Zhao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Jun-Qin Li
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Feng-Ying Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Wei Dong
- Department of Neurology, The Affiliated Yangming Hospital of Ningbo University, Yuyao People's Hospital of Zhejiang Province, Yuyao 315400, Zhejiang, People's Republic of China
| | - Li-Juan Wen
- Institute of Pharmaceutics, College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Wei-Dong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Pei-Lei Yang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Xin-Mei Zhang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| | - Cai-Hong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, People's Republic of China
| |
Collapse
|
41
|
Chen Q, Liang H, Sun Y, Chen Y, He W, Fang X, Sha X, Li J. A carbohydrate mimetic peptide modified size-shrinkable micelle nanocluster for anti-tumor targeting and penetrating drug delivery. Int J Nanomedicine 2019; 14:7339-7352. [PMID: 31686810 PMCID: PMC6751550 DOI: 10.2147/ijn.s213455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/29/2019] [Indexed: 01/02/2023] Open
Abstract
Purpose To deliver the chemotherapeutics through the nanoparticles, the delivery system should accumulate at the tumor site first and then penetrate through the interstitium into the interior. The specific tumor-targeting pathway mediated via the receptor-ligand binding could achieve the desirable accumulation of nanoparticles, and the nanoparticles with smaller sizes were required for penetration. Methods and materials We constructed a size-shrinkable nanocluster modified with a tumor-targeting motif IF-7 (IF-7-MNC) based on a pH-sensitive framework which could be disintegrated in an acid environment to release the micelles aggregated inside. The micelles were constructed by amphiphilic block copolymers PEG−PLA to encapsulate paclitaxel (PTX), while the cross-linked framework consisting of TPGS-PEI was used as a net to gather and release micelles. This nanoplatform could specifically bind with the tumor receptor Annexin A1 through the ligand IF-7 and then shrunk into small micelles with a desirable size for penetration. Conclusion IF-7-MNC of 112.27±6.81 nm could shrink into micelles in PBS (0.01 M, pH 5.0) with sizes of 14.89±0.32 nm. The cellular-uptake results showed that IF-7-MNC could be significantly internalized by A549 cells and HUVEC cells, while the penetration of IF-7-MNC could be more prominent into the 3D-tumor spheroids compared with that of MNC. The biodistribution results displayed that the fluorescence of IF-7-MNC in the tumor site at 24 hrs was 4.5-fold stronger than that of MNC. The results of anti-tumor growth demonstrated that IF-7-MNC was more favorable for the tumor therapy than MNC, where the inhibitory rate of tumor growth was 88.29% in the PTX-loaded IF-7-MNC (IF-7-PMNC) treated group, significantly greater than PMNC treatment group (p<0.05).
Collapse
Affiliation(s)
- Qinyue Chen
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Huihui Liang
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Yali Sun
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Yiting Chen
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Wenxiu He
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Xiaoling Fang
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Xianyi Sha
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, People's Republic of China
| | - Jinming Li
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, People's Republic of China
| |
Collapse
|
42
|
Lu C, Jiang L, Xu W, Yu F, Xia W, Pan M, Zhou W, Pan X, Wu C, Liu D. Poly(ethylene glycol) crosslinked multi-armed poly(ε-benzyloxycarbonyl-L-lysine)s as super-amphiphiles: Synthesis, self-assembly, and evaluation as efficient delivery systems for poorly water-soluble drugs. Colloids Surf B Biointerfaces 2019; 182:110384. [PMID: 31357126 DOI: 10.1016/j.colsurfb.2019.110384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/23/2019] [Accepted: 07/22/2019] [Indexed: 02/05/2023]
Abstract
Polymeric micelles with high thermodynamic stability and loading capacity are of tremendous significance for their potential applications in drug delivery. In the present study, super-amphiphiles in the form of poly(ethylene glycol)-crosslinked multi-armed polyethylenimine-g-poly(ε-benzyloxycarbonyl-L-lysine)s (PEZ-alt-PEG) were designed, synthesized, and optimized as nanocarriers for hydrophobic drugs. In an aqueous solution, the copolymer PEZ-alt-PEG self-assembled into sub-100-nm spherical shell crosslinked micelles with low toxicity in vitro and in vivo. The crosslinked super-amphiphilic structure of PEZ-alt-PEG could not only enhance the thermodynamic stability of polymeric micelles, but it could also significantly improve the loading capacity of hydrophobic drugs, such as curcumin (CUR). CUR-loaded PEZ-alt-PEG micelles could mediate effective drug delivery with sustained and complete CUR release. The use of PEZ-alt-PEG micellar nanocarriers remarkably improved the cellular uptake of CUR and therefore exhibited effective inhibitory activity on the growth of human hepatoma (HepG2) cells. Compared to free CUR, CUR-loaded polymeric micelles significantly accelerated the apoptosis rate of HepG2 cells. Therefore, PEZ-alt-PEG polymeric micelles, with their high thermodynamic stability, high drug-loading capacity, enhanced drug uptake and improved pharmacodynamic effects, could serve as efficient and promising nanocarriers for poorly water-soluble drugs.
Collapse
Affiliation(s)
- Chao Lu
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China; School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, China
| | - Ling Jiang
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Weijie Xu
- Department of Pharmacy, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou 515041, China
| | - Feiyuan Yu
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Wenquan Xia
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Miao Pan
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Wen Zhou
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, China
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, University Town, Guangzhou 510006, China
| | - Daojun Liu
- Shantou University Medical College, 22 Xinling Road, Shantou 515041, China.
| |
Collapse
|
43
|
Wang Y, Xie J, Ai Z, Su J. Nobiletin-loaded micelles reduce ovariectomy-induced bone loss by suppressing osteoclastogenesis. Int J Nanomedicine 2019; 14:7839-7849. [PMID: 31576127 PMCID: PMC6769031 DOI: 10.2147/ijn.s213724] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/30/2019] [Indexed: 01/01/2023] Open
Abstract
Background Nobiletin (NOB), a polymethoxy flavonoid, possesses anti-cancer and anti-inflammatory activities, has been reported that it played role in anti-osteoporosis treatment. However, previous research did not focus on practical use due to lack of hydrophilicity and cytotoxicity at high concentrations. The aim of this study was to develop a therapeutic formulation for osteoporosis based on the utilization of NOB. Methods In this study, NOB-loaded poly(ethylene glycol)-block-poly(e-caprolactone) (NOB-PEG-PCL) was prepared by dialysis method. The effects on osteoclasts and anti-osteoporosis functions were investigated in a RANKL-induced cell model and ovariectomized (OVX) mice. Results Dynamic light scattering and transmission electron microscopy examination results revealed that the NOB-PEG-PCL had a round shape, with a mean diameter around 124 nm. The encapsulation efficiency and drug loading were 76.34±3.25% and 7.60±0.48%, respectively. The in vitro release of NOB from NOB-PEG-PCL showed a remarkably sustained releasing characteristic and could be retained at least 48 hrs in pH 7.4 PBS. Anti-osteoclasts effects demonstrated that the NOB-PEG-PCL significantly inhibited the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear cells stimulated by RANKL. Furthermore, the NOB-PEG-PCL did not produce cytotoxicity on bone marrow-derived macrophages (BMMs). The mRNA expressions of genetic markers of osteoclasts including TRAP and cathepsin K were significantly decreased in the presence of NOB-PEG-PCL. In addition, the NOB-PEG-PCL inhibited OC differentiation of BMMs through RANKL-induced MAPK signal pathway. After administration of the NOB-PEG-PCL, NOB-PEG-PCL prevented bone loss and improved bone density in OVX mice. These findings suggest that NOB-PEG-PCL might have great potential in the treatment of osteoporosis. Conclusion The results suggested that NOB-PEG-PCL micelles could effectively prevent NOB fast release from micelles and extend circulation time. The NOB-PEG-PCL delivery system may be a promising way to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Yabing Wang
- Department of Prosthodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, People's Republic of China
| | - Jian Xie
- Department of Prosthodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, People's Republic of China
| | - Zexin Ai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, People's Republic of China
| | - Jiansheng Su
- Department of Prosthodontics, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, People's Republic of China
| |
Collapse
|
44
|
Karabasz A, Lachowicz D, Karewicz A, Mezyk-Kopec R, Stalińska K, Werner E, Cierniak A, Dyduch G, Bereta J, Bzowska M. Analysis of toxicity and anticancer activity of micelles of sodium alginate-curcumin. Int J Nanomedicine 2019; 14:7249-7262. [PMID: 31564877 PMCID: PMC6735652 DOI: 10.2147/ijn.s213942] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
Background Curcumin is a natural polyphenol with anti-inflammatory, chemopreventive and anticancer activity. However, its high hydrophobicity and poor bioavailability limit its medical application. The development of nanocarriers for curcumin delivery is an attractive approach to overcome its low bioavailability and fast metabolism in the liver. We synthesized a blood compatible alginate-curcumin conjugate, AA-Cur, which formed colloidally stable micelles of approximately 200 nm and, as previously shown, exerted strong cytotoxicity against mouse cancer cell lines. Here we analyze in vivo toxicity and antitumor activity of AA-Cur in two different mouse tumor models. Method Potential toxicity of intravenously injected AA-Cur was evaluated by: i) analyses of blood parameters (morphology and biochemistry), ii) histology, iii) DNA integrity (comet assay), and iv) cytokine profiling (flow cytometry). Antitumor activity of AA-Cur was evaluated by measuring the growth of subcutaneously inoculated colon MC38-CEA- or orthotopically injected breast 4T1 tumor cells in control mice vs mice treated with AA-Cur. Results Injections of four doses of AA-Cur did not reveal any toxicity of the conjugate, thus indicating the safety of its use. AA-Cur elicited moderate anti-tumor activity toward colon MC38-CEA or breast 4T1 carcinomas. Conclusion The tested conjugate of alginate and curcumin, AA-Cur, is non-toxic and safe, but exhibits limited anticancer activity.
Collapse
Affiliation(s)
- Alicja Karabasz
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dorota Lachowicz
- Academic Centre for Materials and Nanotechnology, AGH University of Science and Technology, Kraków, Poland
| | - Anna Karewicz
- Faculty of Chemistry, Jagiellonian University, Kraków, Poland
| | - Renata Mezyk-Kopec
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krystyna Stalińska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.,Department of Animal Reproduction and Anatomy, Faculty of Animal Science, University of Agriculture, Krakow, Poland
| | - Agnieszka Cierniak
- Department of General Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Grzegorz Dyduch
- Department of Pathomorphology, Jagiellonian University Medical College, Kraków, Poland
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Monika Bzowska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
45
|
Core-shell-corona chitosan-based micelles for tumor intracellular pH-triggered drug delivery: Improving performance by grafting polycation. Int J Biol Macromol 2019; 141:161-170. [PMID: 31479675 DOI: 10.1016/j.ijbiomac.2019.08.251] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 02/01/2023]
Abstract
Core-shell-corona chitosan-based micelles were designed for the tumor intracellular pH-triggered doxorubicin (DOX) delivery, via a facile in-situ micellization in an aqueous solution of DOX and polyethylene glycol (PEG) and poly(2-(diisopropylamino) ethyl methacrylate) (PDPA) dual-modified chitosan (PEG-g-CS-g-PDPA). The effect of the PDPA modification on the diameter, drug loading-capacity (DLC) and pH-triggered drug release was investigated for the three different polymerization degrees of PDPA (25, 32, and 42) with a similar modification degree of ~22%. The optimized ones, the core-shell-corona DOX/PEG-g-CS-g-PDPA32 micelles possessed a mean hydrodynamic diameter (Dh) of 211 nm and DLC of 54%, showing an excellent pH-triggered drug release with negligible premature drug leakage in 60 h. Such results indicated that grafting polycation could efficiently improve the performance of the chitosan-based drug delivery system (DDS) for tumor chemotherapy.
Collapse
|
46
|
Zhou X, Wu X, Chen B. Sorcin: a novel potential target in therapies of cancers. Cancer Manag Res 2019; 11:7327-7336. [PMID: 31496794 PMCID: PMC6689139 DOI: 10.2147/cmar.s208677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/05/2019] [Indexed: 12/14/2022] Open
Abstract
Soluble resistance-related calcium-binding protein (sorcin) is a member of the penta-EF-hand protein family. Sorcin is widely distributed in normal human tissues, such as the brain, heart, lymphocytes, kidneys, breast and skin. Findings suggest that sorcin is associated with the regulation of calcium homeostasis, cell cycle and vesicle trafficking. It has been reported that many types of non-neoplastic diseases such as diabetes, viral infection, infertility, and nervous system diseases were affected by the expression of sorcin. One of the main issues is the role of sorcin in neoplastic diseases. Research proved that sorcin can be found to overexpress in cells of several cancers, particularly in the case of multidrug-resistant cancers. Additionally, the researchers proposed that the expression of sorcin was significantly associated with the foundation of multidrug resistance (MDR). All the findings mentioned above emphasized the importance of studying sorcin. This review mainly includes the following aspects: functions of sorcin, role in non-neoplastic and neoplastic diseases, and research related to drugs. To sum up, sorcin is a potential novel target to be studied to deal with MDR.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Xue Wu
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
47
|
Formulation of Nanomicelles to Improve the Solubility and the Oral Absorption of Silymarin. Molecules 2019; 24:molecules24091688. [PMID: 31052197 PMCID: PMC6540123 DOI: 10.3390/molecules24091688] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 01/13/2023] Open
Abstract
Two novel nanomicellar formulations were developed to improve the poor aqueous solubility and the oral absorption of silymarin. Polymeric nanomicelles made of Soluplus and mixed nanomicelles combining Soluplus with d-α-tocopherol polyethylene glycol 1000 succinate (vitamin E TPGS) were prepared using the thin film method. Physicochemical parameters were investigated, in particular the average diameter, the homogeneity (expressed as polydispersity index), the zeta potential, the morphology, the encapsulation efficiency, the drug loading, the critical micellar concentration and the cloud point. The sizes of ~60 nm, the narrow size distribution (polydispersity index ≤0.1) and the encapsulation efficiency >92% indicated the high affinity between silymarin and the core of the nanomicelles. Solubility studies demonstrated that the solubility of silymarin increased by ~6-fold when loaded into nanomicelles. Furthermore, the physical and chemical parameters of SLM-loaded formulations stored at room temperature and in refrigerated conditions (4 °C) were monitored over three months. In vitro stability and release studies in media miming the physiological conditions were also performed. In addition, both formulations did not alter the antioxidant properties of silymarin as evidenced by the 1,1-Diphenyl-2-picrylhydrazyl radical (DPPH) assay. The potential of the nanomicelles to increase the intestinal absorption of silymarin was firstly investigated by the parallel artificial membrane permeability assay. Subsequently, transport studies employing Caco-2 cell line demonstrated that mixed nanomicelles statistically enhanced the permeability of silymarin compared to polymeric nanomicelles and unformulated extract. Finally, the uptake studies indicated that both nanomicellar formulations entered into Caco-2 cells via energy-dependent mechanisms.
Collapse
|
48
|
Mishra H, Mishra PK, Iqbal Z, Jaggi M, Madaan A, Bhuyan K, Gupta N, Gupta N, Vats K, Verma R, Talegaonkar S. Co-Delivery of Eugenol and Dacarbazine by Hyaluronic Acid-Coated Liposomes for Targeted Inhibition of Survivin in Treatment of Resistant Metastatic Melanoma. Pharmaceutics 2019; 11:E163. [PMID: 30987266 PMCID: PMC6523131 DOI: 10.3390/pharmaceutics11040163] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
While melanoma remains a challenge for oncologists, possibilities are being continuously explored to fight resistant metastatic melanoma more effectively. Eugenol is reported to inhibit survivin protein in breast cancer cells. Survivin is also overexpressed by melanoma cells, and is known to impart resistance to them against chemotherapy-induced apoptosis. To be able to fight resistant melanoma, we formulated hyaluronic acid (HA)-coated liposomes loaded with an effective combination of anti-melanoma agents (Dacarbazine and Eugenol), using a solvent injection method. Quality-by-Design (QbD) was applied to optimize and obtain a final formulation with the desired quality attributes, and within an acceptable size range. The optimized formulation was then subjected to performance analysis in cell lines. Coated-Dacarbazine Eugenol Liposomes were found to possess 95.08% cytotoxicity at a dacarbazine concentration of 0.5 µg/mL, while Dacarbazine Solution showed only 10.20% cytotoxicity at the same concentration. The number of late apoptotic cells was also found to be much higher (45.16% vs. 8.43%). Furthermore, migration assay and proliferation study also revealed significantly higher inhibition of cell migration and proliferation by Coated-Dacarbazine Eugenol Liposomes, signifying its potential against metastasis. Thus, surface-functionalized dacarbazine- and eugenol-loaded liposomes hold great promise against resistant and aggressive metastatic melanoma, with much less unwanted cytotoxicity and reduced doses of the chemotherapeutic agent.
Collapse
Affiliation(s)
- Harshita Mishra
- Departmant of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Pawan Kumar Mishra
- Department of Wood Processing, Mendel University in Brno, 61300 Brno, Czech Republic.
| | - Zeenat Iqbal
- Departmant of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Manu Jaggi
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Alka Madaan
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Kimi Bhuyan
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Namita Gupta
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Neha Gupta
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Karnika Vats
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Ritu Verma
- Dabur Research Foundation, Ghaziabad 201010, India.
| | - Sushama Talegaonkar
- Departmant of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Govt. of NCT of Delhi, Pushp Vihar, New Delhi 110017, India.
| |
Collapse
|
49
|
Transforming an inert nanopolymer into broad-spectrum bactericidal by superstructure tuning. Colloids Surf B Biointerfaces 2019; 178:214-221. [PMID: 30870788 DOI: 10.1016/j.colsurfb.2019.02.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 01/19/2023]
Abstract
Poloxamer block copolymers (also known as Pluronic®) are particularly useful for drug delivery and self-assembly techniques. These nanopolymers are generally considered to be biologically inert and they were used to generate only bacteria repellent surfaces but keeps bacteria alive and as a latent threat. However, the inherent capabilities of these nanopolymers to kill bacteria have been largely overlooked. Here, we report that Pluronic shaped as superstructures (self-organized array of micelles) in fact possess a broad-spectrum bactericidal activity (capability of killing bacteria) similar to that shown for some antibiotics. This further represents the first report that shows that appropriate control of superstructured mesophase architecture is a key parameter for bactericidal efficacy. Based on this finding, we have developed a highly bactericidal coating (>99.9% kill) against all tested Gram-positive (Staphylococcus aureus and Bacillus subtilis) and Gram-negative (Salmonella typhimurium LT2, Escherichia coli K12 and Pseudomonas aeruginosa PAO1) bacteria which moreover allows the adhesion and proliferation of mammalian cells. The inexpensiveness and ease of production make these versatile nanopolymer structures a powerful tool for the development of a new generation of highly effective antimicrobial coatings.
Collapse
|
50
|
Limonta P, Moretti RM, Marzagalli M, Fontana F, Raimondi M, Montagnani Marelli M. Role of Endoplasmic Reticulum Stress in the Anticancer Activity of Natural Compounds. Int J Mol Sci 2019; 20:ijms20040961. [PMID: 30813301 PMCID: PMC6412802 DOI: 10.3390/ijms20040961] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/04/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer represents a serious global health problem, and its incidence and mortality are rapidly growing worldwide. One of the main causes of the failure of an anticancer treatment is the development of drug resistance by cancer cells. Therefore, it is necessary to develop new drugs characterized by better pharmacological and toxicological profiles. Natural compounds can represent an optimal collection of bioactive molecules. Many natural compounds have been proven to possess anticancer effects in different types of tumors, but often the molecular mechanisms associated with their cytotoxicity are not completely understood. The endoplasmic reticulum (ER) is an organelle involved in multiple cellular processes. Alteration of ER homeostasis and its appropriate functioning originates a cascade of signaling events known as ER stress response or unfolded protein response (UPR). The UPR pathways involve three different sensors (protein kinase RNA(PKR)-like ER kinase (PERK), inositol requiring enzyme1α (IRE1) and activating transcription factor 6 (ATF6)) residing on the ER membranes. Although the main purpose of UPR is to restore this organelle's homeostasis, a persistent UPR can trigger cell death pathways such as apoptosis. There is a growing body of evidence showing that ER stress may play a role in the cytotoxicity of many natural compounds. In this review we present an overview of different plant-derived natural compounds, such as curcumin, resveratrol, green tea polyphenols, tocotrienols, and garcinia derivates, that exert their anticancer activity via ER stress modulation in different human cancers.
Collapse
Affiliation(s)
- Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| | - Roberta M Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|