51
|
Borzęcka W, Pereira P, Fernandes R, Trindade T, Torres T, Tome J. Spherical and rod shaped mesoporous nanosilicas for cancer-targeted and photosensitizers delivery in photodynamic therapy. J Mater Chem B 2022; 10:3248-3259. [DOI: 10.1039/d1tb02299g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesoporous silica nanoparticles (MSNPs) have attracted much attention in many biomedical applications. One of the fields in which smart functional nanosystems have found wide application is in cancer treatment. Here,...
Collapse
|
52
|
Gupta P, Neupane YR, Parvez S, Kohli K. Recent advances in targeted nanotherapeutic approaches for breast cancer management. Nanomedicine (Lond) 2021; 16:2605-2631. [PMID: 34854336 DOI: 10.2217/nnm-2021-0281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly occurring tumor disease worldwide. Breast cancer is currently managed by conventional chemotherapy, which is inadequate in curbing this heterogeneous disease and results in off-site toxic effects, suggesting effective treatment approaches with better therapeutic profiles are needed. This review, therefore, focuses on the recent advancements in delivering therapeutics to the target site using passive and/or active targeted nanodrug-delivery systems to ameliorate endolysosomal escape. In addition, recent strategies in targeting breast cancer stem cells are discussed. The role of naturally cell-secreted nanovesicles (exosomes) in the management of triple-negative breast cancer is also discussed.
Collapse
Affiliation(s)
- Priya Gupta
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yub Raj Neupane
- Department of Pharmacy, National University of Singapore, Singapore, 117559
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.,Lloyd Institute of Management & Technology (Pharm.), Plot No. 11, Knowledge Park-II, Greater Noida, 201308, Uttar Pradesh, India
| |
Collapse
|
53
|
Yap KM, Sekar M, Fuloria S, Wu YS, Gan SH, Mat Rani NNI, Subramaniyan V, Kokare C, Lum PT, Begum MY, Mani S, Meenakshi DU, Sathasivam KV, Fuloria NK. Drug Delivery of Natural Products Through Nanocarriers for Effective Breast Cancer Therapy: A Comprehensive Review of Literature. Int J Nanomedicine 2021; 16:7891-7941. [PMID: 34880614 PMCID: PMC8648329 DOI: 10.2147/ijn.s328135] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Despite recent advances in the diagnosis and treatment of breast cancer (BC), it remains a global health issue affecting millions of women annually. Poor prognosis in BC patients is often linked to drug resistance as well as the lack of effective therapeutic options for metastatic and triple-negative BC. In response to these unmet needs, extensive research efforts have been devoted to exploring the anti-BC potentials of natural products owing to their multi-target mechanisms of action and good safety profiles. Various medicinal plant extracts/essential oils and natural bioactive compounds have demonstrated anti-cancer activities in preclinical BC models. Despite the promising preclinical results, however, the clinical translation of natural products has often been hindered by their poor stability, aqueous solubility and bioavailability. There have been attempts to overcome these limitations, particularly via the use of nano-based drug delivery systems (NDDSs). This review highlights the tumour targeting mechanisms of NDDSs, the advantages and disadvantages of the major classes of NDDSs and their current clinical status in BC treatment. Besides, it also discusses the proposed anti-BC mechanisms and nanoformulations of nine medicinal plants' extracts/essential oils and nine natural bioactive compounds; selected via the screening of various scientific databases, including PubMed, Scopus and Google Scholar, based on the following keywords: "Natural Product AND Nanoparticle AND Breast Cancer". Overall, these nanoformulations exhibit improved anti-cancer efficacy against preclinical BC models, with some demonstrating biocompatibility with normal cell lines and mouse models. Further clinical studies are, however, warranted to ascertain their efficacy and biocompatibility in humans.
Collapse
Affiliation(s)
- Kah Min Yap
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | | | - Yuan Seng Wu
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Selangor, 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor, 47500, Malaysia
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Nur Najihah Izzati Mat Rani
- Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | | | - Chandrakant Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society’s, Sinhgad Institute of Pharmacy, Narhe, Pune, 411041, India
| | - Pei Teng Lum
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University (KKU), Asir-Abha, 61421, Saudi Arabia
| | - Shankar Mani
- Department of Pharmaceutical Chemistry, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, Mandya, Karnataka, 571418, India
| | | | | | | |
Collapse
|
54
|
Lin MHC, Chang LC, Chung CY, Huang WC, Lee MH, Chen KT, Lai PS, Yang JT. Photochemical Internalization of Etoposide Using Dendrimer Nanospheres Loaded with Etoposide and Protoporphyrin IX on a Glioblastoma Cell Line. Pharmaceutics 2021; 13:pharmaceutics13111877. [PMID: 34834292 PMCID: PMC8621426 DOI: 10.3390/pharmaceutics13111877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary neoplasm of the adult central nervous system originating from glial cells. The prognosis of those affected by GBM has remained poor despite advances in surgery, chemotherapy, and radiotherapy. Photochemical internalization (PCI) is a release mechanism of endocytosed therapeutics into the cytoplasm, which relies on the membrane disruptive effect of light-activated photosensitizers. In this study, phototherapy by PCI was performed on a human GBM cell-line using the topoisomerase II inhibitor etoposide (Etop) and the photosensitizer protoporphyrin IX (PpIX) loaded in nanospheres (Ns) made from generation-5 polyamidoamine dendrimers (PAMAM(G5)). The resultant formulation, Etop/PpIX-PAMAM(G5) Ns, measured 217.4 ± 2.9 nm in diameter and 40.5 ± 1.3 mV in charge. Confocal microscopy demonstrated PpIX fluorescence within the endo-lysosomal compartment, and an almost twofold increase in cellular uptake compared to free PpIX by flow cytometry. Phototherapy with 3 min and 5 min light illumination resulted in a greater extent of synergism than with co-administered Etop and PpIX; notably, antagonism was observed without light illumination. Mechanistically, significant increases in oxidative stress and apoptosis were observed with Etop/PpIX-PAMAM(G5) Ns upon 5 min of light illumination in comparison to treatment with either of the agents alone. In conclusion, simultaneous delivery and endo-lysosomal co-localization of Etop and PpIX by PAMAM(G5) Ns leads to a synergistic effect by phototherapy; in addition, the finding of antagonism without light illumination can be advantageous in lowering the dark toxicity and improving photo-selectivity.
Collapse
Affiliation(s)
- Martin Hsiu-Chu Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan;
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Li-Ching Chang
- Department of Dentistry, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan;
- Department of Nursing, Chang Gung University of Science and Technology, Chia-Yi 61363, Taiwan
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Wei-Chao Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Ming-Hsueh Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Kuo-Tai Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan;
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (C.-Y.C.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
- Correspondence: ; Tel.: +886-5-3621000 (ext. 3412); Fax: +886-5-3621000 (ext. 3002)
| |
Collapse
|
55
|
Li X, Yu L, Zhang C, Niu X, Sun M, Yan Z, Wang W, Yuan Z. Tumor acid microenvironment-activated self-targeting & splitting gold nanoassembly for tumor chemo-radiotherapy. Bioact Mater 2021; 7:377-388. [PMID: 34466739 PMCID: PMC8379383 DOI: 10.1016/j.bioactmat.2021.05.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022] Open
Abstract
Low accumulation and penetration of nanomedicines in tumor severely reduce therapeutic efficacy. Herein, a pH-responsive gold nanoassembly is designed to overcome these problems. Polyethylene glycol linked raltitrexed (RTX, target ligand and chemotherapy drug) and two tertiary amine molecules (1-(2-aminoethyl) pyrrolidine and N,N-dibutylethylenediamine) are modified on the surface of the 6-nm gold nanoparticles by lipoic acid to form gold nanoassembly defined as Au-NNP(RTX). The Au-NNP (RTX) nanoassembly could remain at about 160 nm at the blood circulation (pH 7.4), while split into 6-nm gold nanoparticles due to tertiary amine protonation at tumor extracellular pH (pH 6.8). This pH-responsive disassembly behavior endows Au-NNP(RTX) better tumor tissue permeability through the better diffusion brought by the size reduction. Meanwhile, after disassembly, more RTXs on the surface of gold nanoparticles are exposed from the shielded state of assembly along with 2.25-fold augment of cellular uptake capability. Most importantly, the results show that Au-NNP(RTX) possesses of high tumor accumulation and effective tumor penetration, thereby enhancing the tumor chemo-radiotherapy efficiency. A pH-responsive self-targeting & splitting gold nanoassembly is fabricated. The nanoassembly holds better tumor tissue permeability by the size reduction. The nanoassembly enhances targeting capability by ligand shielding and exposure. Clever design endows the system synergistic effect of chemo-radiotherapy.
Collapse
Affiliation(s)
- Xiaomin Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Licheng Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Xiaoyan Niu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Mengjie Sun
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zichao Yan
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wei Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhi Yuan
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071, China.,Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| |
Collapse
|
56
|
Kankala RK, Xu PY, Chen BQ, Wang SB, Chen AZ. Supercritical fluid (SCF)-assisted fabrication of carrier-free drugs: An eco-friendly welcome to active pharmaceutical ingredients (APIs). Adv Drug Deliv Rev 2021; 176:113846. [PMID: 34197896 DOI: 10.1016/j.addr.2021.113846] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 02/09/2023]
Abstract
Despite the success in developing various pharmaceutical formulations, most of the active pharmaceutical ingredients (APIs)/drugs, according to the Biopharmaceutics Classification System (BCS), often suffer from various intrinsic limitations of solubility and permeability, substantially hindering their bioavailability in vivo. Regardless of the fact that the availability of different particle fabrication approaches (top-down and bottom-up) towards pharmaceutical manufacturing, the supercritical fluid (SCF) technology has emerged as one of the highly effective substitutes due to the environmentally benign nature and processing convenience, as well as the economically promising character of SCFs. The exceptional features of SCFs have endowed the fabrication of various APIs either solely or in combination with the compatible supramolecular species towards achieving improved drug delivery. Operating such APIs in high-pressure conditions often results in arbitrary-sized particulate forms, ranging from micron-sized to sub-micron/nano-sized particles. Comparatively, these SCF-processed particles offer enhanced tailorable physicochemical and morphological properties (size, shape, and surface), as well as improved performance efficacy (bioavailability and therapy) over the unprocessed APIs. Although the "carrier-based" delivery is practical among diverse delivery systems, the direct fabrication of APIs into suitable particulate forms, referred to as "carrier-free" delivery, has increased attention towards improving the bioavailability and conveying a high payload of the APIs. This review gives a comprehensive emphasis on the SCF-assisted fabrication of diverse APIs towards exploring their great potential in drug delivery. Initially, we discuss various challenges of drug delivery and particle fabrication approaches. Further, different supercritical carbon dioxide (SC-CO2)-based fabrication approaches depending on the character of SCFs are explicitly described, highlighting their advantages and suitability in processing diverse APIs. Then, we provide detailed insights on various processing factors affecting the properties and morphology of SCF-processed APIs and their pharmaceutical applications, emphasizing their performance efficacy when administered through multiple routes of administration. Finally, we summarize this compilation with exciting perspectives based on the lessons learned so far and moving forward in terms of challenges and opportunities in the scale-up and clinical translation of these drugs using this innovative technology.
Collapse
|
57
|
Zhang M, Xu N, Xu W, Ling G, Zhang P. Potential therapies and diagnosis based on Golgi-targeted nano drug delivery systems. Pharmacol Res 2021; 175:105861. [PMID: 34464677 DOI: 10.1016/j.phrs.2021.105861] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
With the rapid development of nanotechnology, organelle-targeted nano drug delivery systems (NDDSs) have emerged as a potential method which can transport drugs specifically to the subcellular compartments like nucleus, mitochondrion, lysosome, endoplasmic reticulum (ER) and Golgi apparatus (GA). GA not only plays a key role in receiving, modifying, packaging and transporting proteins and lipids, but also contributes to a set of cellular processes. Golgi-targeted NDDSs can alter the morphology of GA and will become a promising strategy with high specificity, low-dose administration and decreased occurrence of side effects. In this review, Golgi-targeted NDDSs and their applications in disease therapies and diagnosis such as cancer, metastasis, fibrosis and neurological diseases are introduced. Meanwhile, modifications of NDDSs to achieve targeting strategies, Golgi-disturbing agents to change the morphology of GA, special endocytosis to achieve endosomal/lysosomal escape strategies are also involved.
Collapse
Affiliation(s)
- Manyue Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Na Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wenxin Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
58
|
Wang P, Kankala RK, Chen B, Zhang Y, Zhu M, Li X, Long R, Yang D, Krastev R, Wang S, Xiong X, Liu Y. Cancer Cytomembrane-Cloaked Prussian Blue Nanoparticles Enhance the Efficacy of Mild-Temperature Photothermal Therapy by Disrupting Mitochondrial Functions of Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:37563-37577. [PMID: 34338525 DOI: 10.1021/acsami.1c11138] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Despite its success against cancer, photothermal therapy (PTT) (>50 °C) suffers from several limitations such as triggering inflammation and facilitating immune escape and metastasis and also damage to the surrounding normal cells. Mild-temperature PTT has been proposed to override these shortcomings. We developed a nanosystem using HepG2 cancer cell membrane-cloaked zinc glutamate-modified Prussian blue nanoparticles with triphenylphosphine-conjugated lonidamine (HmPGTL NPs). This innovative approach achieved an efficient mild-temperature PTT effect by downregulating the production of intracellular ATP. This disrupts a section of heat shock proteins that cushion cancer cells against heat. The physicochemical properties, anti-tumor efficacy, and mechanisms of HmPGTL NPs both in vitro and in vivo were investigated. Moreover, the nanoparticles cloaked with the HepG2 cell membrane substantially prolonged the circulation time in vivo. Overall, the designed nanocomposites enhance the efficacy of mild-temperature PTT by disrupting the production of ATP in cancer cells. Thus, we anticipate that the mild-temperature PTT nanosystem will certainly present its enormous potential in various biomedical applications.
Collapse
Affiliation(s)
- Pei Wang
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Jiangxi Key Laboratory of Stomatology and Biomedicine, School of Stomatology, Nanchang University, Nanchang 330006, P. R. China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Biaoqi Chen
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361021, P. R. China
| | - Mingzhi Zhu
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Xuemei Li
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ruimin Long
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Dayun Yang
- Institute for Translational Medicine, School of Basic Medical Science, Fujian Medical University, Fuzhou 350122, P. R. China
| | - Rumen Krastev
- Faculty for Applied Chemistry, Reutlingen University, Alteburgstr, 150, Reutlingen 72762, Germany
| | - Shibin Wang
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Xin Xiong
- NMI Natural and Medical Sciences Institute, University of Tübingen, Markwiesenstr, 55, Reutlingen 72770, Germany
| | - Yuangang Liu
- Fujian Provincial Key Laboratory of Biochemical Technology, Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| |
Collapse
|
59
|
Tremi I, Spyratou E, Souli M, Efstathopoulos EP, Makropoulou M, Georgakilas AG, Sihver L. Requirements for Designing an Effective Metallic Nanoparticle (NP)-Boosted Radiation Therapy (RT). Cancers (Basel) 2021; 13:cancers13133185. [PMID: 34202342 PMCID: PMC8269428 DOI: 10.3390/cancers13133185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent advances in nanotechnology gave rise to trials with various types of metallic nanoparticles (NPs) to enhance the radiosensitization of cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy. This work reviews the physical and chemical mechanisms leading to the enhancement of ionizing radiation’s detrimental effects on cells and tissues, as well as the plethora of experimental procedures to study these effects of the so-called “NPs’ radiosensitization”. The paper presents the need to a better understanding of all the phases of actions before applying metallic-based NPs in clinical practice to improve the effect of IR therapy. More physical and biological experiments especially in vivo must be performed and simulation Monte Carlo or mathematical codes based on more accurate models for all phases must be developed. Abstract Many different tumor-targeted strategies are under development worldwide to limit the side effects and improve the effectiveness of cancer therapies. One promising method is to enhance the radiosensitization of the cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy using metallic nanoparticles (NPs). Radiotherapy with MV photons is more commonly available and applied in cancer clinics than high LET particle radiotherapy, so the addition of high-Z NPs has the potential to further increase the efficacy of photon radiotherapy in terms of NP radiosensitization. Generally, when using X-rays, mainly the inner electron shells are ionized, which creates cascades of both low and high energy Auger electrons. When using high LET particles, mainly the outer shells are ionized, which give electrons with lower energies than when using X-rays. The amount of the produced low energy electrons is higher when exposing NPs to heavy charged particles than when exposing them to X-rays. Since ions traverse the material along tracks, and therefore give rise to a much more inhomogeneous dose distributions than X-rays, there might be a need to introduce a higher number of NPs when using ions compared to when using X-rays to create enough primary and secondary electrons to get the desired dose escalations. This raises the questions of toxicity. This paper provides a review of the fundamental processes controlling the outcome of metallic NP-boosted photon beam and ion beam radiation therapy and presents some experimental procedures to study the biological effects of NPs’ radiosensitization. The overview shows the need for more systematic studies of the behavior of NPs when exposed to different kinds of ionizing radiation before applying metallic-based NPs in clinical practice to improve the effect of IR therapy.
Collapse
Affiliation(s)
- Ioanna Tremi
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Maria Souli
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Mersini Makropoulou
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Correspondence: (A.G.G.); (L.S.)
| | - Lembit Sihver
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Correspondence: (A.G.G.); (L.S.)
| |
Collapse
|
60
|
Clement S, Anwer AG, Pires L, Campbell J, Wilson BC, Goldys EM. Radiodynamic Therapy Using TAT Peptide-Targeted Verteporfin-Encapsulated PLGA Nanoparticles. Int J Mol Sci 2021; 22:ijms22126425. [PMID: 34204001 PMCID: PMC8232618 DOI: 10.3390/ijms22126425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022] Open
Abstract
Radiodynamic therapy (RDT) is a recent extension of conventional photodynamic therapy, in which visible/near infrared light irradiation is replaced by a well-tolerated dose of high-energy X-rays. This enables greater tissue penetration to allow non-invasive treatment of large, deep-seated tumors. We report here the design and testing of a drug delivery system for RDT that is intended to enhance intra- or peri-nuclear localization of the photosensitizer, leading to DNA damage and resulting clonogenic cell kill. This comprises a photosensitizer (Verteporfin, VP) incorporated into poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) that are surface-functionalized with a cell-penetrating HIV trans-activator of transcription (TAT) peptide. In addition to a series of physical and photophysical characterization studies, cytotoxicity tests in pancreatic (PANC-1) cancer cells in vitro under 4 Gy X-ray exposure from a clinical 6 MV linear accelerator (LINAC) showed that TAT targeting of the nanoparticles markedly enhances the effectiveness of RDT treatment, particularly when assessed by a clonogenic, i.e., DNA damage-mediated, cell kill.
Collapse
Affiliation(s)
- Sandhya Clement
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
- Correspondence:
| | - Ayad G. Anwer
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| | - Layla Pires
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada; (L.P.); (B.C.W.)
| | - Jared Campbell
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| | - Brian C. Wilson
- Princess Margaret Cancer Centre, University Health Network and Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada; (L.P.); (B.C.W.)
| | - Ewa M. Goldys
- ARC Centre of Excellence in Nanoscale Biophotonics, The Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; (A.G.A.); (J.C.); (E.M.G.)
- ARC Centre of Excellence in Nanoscale Biophotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
61
|
S Allemailem K, Almatroudi A, Alsahli MA, Aljaghwani A, M El-Kady A, Rahmani AH, Khan AA. Novel Strategies for Disrupting Cancer-Cell Functions with Mitochondria-Targeted Antitumor Drug-Loaded Nanoformulations. Int J Nanomedicine 2021; 16:3907-3936. [PMID: 34135584 PMCID: PMC8200140 DOI: 10.2147/ijn.s303832] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/24/2021] [Indexed: 12/16/2022] Open
Abstract
Any variation in normal cellular function results in mitochondrial dysregulation that occurs in several diseases, including cancer. Such processes as oxidative stress, metabolism, signaling, and biogenesis play significant roles in cancer initiation and progression. Due to their central role in cellular metabolism, mitochondria are favorable therapeutic targets for the prevention and treatment of conditions like neurodegenerative diseases, diabetes, and cancer. Subcellular mitochondria-specific theranostic nanoformulations for simultaneous targeting, drug delivery, and imaging of these organelles are of immense interest in cancer therapy. It is a challenging task to cross multiple barriers to target mitochondria in diseased cells. To overcome these multiple barriers, several mitochondriotropic nanoformulations have been engineered for the transportation of mitochondria-specific drugs. These nanoformulations include liposomes, dendrimers, carbon nanotubes, polymeric nanoparticles (NPs), and inorganic NPs. These nanoformulations are made mitochondriotropic by conjugating them with moieties like dequalinium, Mito-Porter, triphenylphosphonium, and Mitochondria-penetrating peptides. Most of these nanoformulations are meticulously tailored to control their size, charge, shape, mitochondriotropic drug loading, and specific cell-membrane interactions. Recently, some novel mitochondria-selective antitumor compounds known as mitocans have shown high toxicity against cancer cells. These selective compounds form vicious oxidative stress and reactive oxygen species cycles within cancer cells and ultimately push them to cell death. Nanoformulations approved by the FDA and EMA for clinical applications in cancer patients include Doxil, NK105, and Abraxane. The novel use of these NPs still faces tremendous challenges and an immense amount of research is needed to understand the proper mechanisms of cancer progression and control by these NPs. Here in this review, we summarize current advancements and novel strategies of delivering different anticancer therapeutic agents to mitochondria with the help of various nanoformulations.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Aseel Aljaghwani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Asmaa M El-Kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
62
|
Oladimeji O, Akinyelu J, Daniels A, Singh M. Modified Gold Nanoparticles for Efficient Delivery of Betulinic Acid to Cancer Cell Mitochondria. Int J Mol Sci 2021; 22:ijms22105072. [PMID: 34064888 PMCID: PMC8150271 DOI: 10.3390/ijms22105072] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023] Open
Abstract
Advances in nanomedicine have seen the adaptation of nanoparticles (NPs) for subcellular delivery for enhanced therapeutic impact and reduced side effects. The pivotal role of the mitochondria in apoptosis and their potential as a target in cancers enables selective induction of cancer cell death. In this study, we examined the mitochondrial targeted delivery of betulinic acid (BA) by the mitochondriotropic TPP+-functionalized epigallocatechin gallate (EGCG)-capped gold NPs (AuNPs), comparing the impact of polyethylene glycol (PEG) and poly-L-lysine-graft-polyethylene glycol (PLL-g-PEG) copolymer on delivery efficacy. This included the assessment of their cellular uptake, mitochondrial localization and efficacy as therapeutic delivery platforms for BA in the human Caco-2, HeLa and MCF-7 cancer cell lines. These mitochondrial-targeted nanocomplexes demonstrated significant inhibition of cancer cell growth, with targeted nanocomplexes recording IC50 values in the range of 3.12–13.2 µM compared to that of the free BA (9.74–36.31 µM) in vitro, demonstrating the merit of mitochondrial targeting. Their mechanisms of action implicated high amplitude mitochondrial depolarization, caspases 3/7 activation, with an associated arrest at the G0/G1 phase of the cell cycle. This nano-delivery system is a potentially viable platform for mitochondrial-targeted delivery of BA and highlights mitochondrial targeting as an option in cancer therapy.
Collapse
|
63
|
Niu S, Zhang X, Williams GR, Wu J, Gao F, Fu Z, Chen X, Lu S, Zhu LM. Hollow Mesoporous Silica Nanoparticles Gated by Chitosan-Copper Sulfide Composites as Theranostic Agents for the Treatment of Breast Cancer. Acta Biomater 2021; 126:408-420. [PMID: 33731303 DOI: 10.1016/j.actbio.2021.03.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/19/2022]
Abstract
The combination of chemotherapy and photothermal therapy (PTT) into a single formulation has attracted increasing attention as a strategy for enhancing cancer treatment. Here, hollow mesoporous silica nanoparticles (HMSNs) were used as a base carrier material, loaded with the anti-cancer drug doxorubicin (DOX), and surface functionalized with chitosan (CS) and copper sulfide (CuS) nanodots to give HMSNs-CS-DOX@CuS. In this formulation, the CuS dots act as gatekeepers to seal the surface pores of the HMSNs, preventing a burst release of DOX into the systemic circulation. S-S bonds connect the CuS dots to the HMSNs; these are selectively cleaved under the reducing microenvironment of the tumor, permitting targeted drug release. This, coupled with the PTT properties of CuS, results in a potent chemo/PTT platform. The HMSNs-CS-DOX@CuS nanoparticles have a uniform size (150 ± 13 nm), potent photothermal properties (η = 36.4 %), and tumor-targeted and near infrared (NIR) laser irradiation-triggered DOX release. In vitro and in vivo experimental results confirmed that the material has good biocompatibility, but is effectively taken up by cancer cells. Moreover, the CuS nanodots permit simultaneous thermal/photoacoustic dual-modality imaging. Treatment with HMSNs-CS-DOX@CuS and NIR irradiation caused extensive apoptosis in cancer cells both in vitro and in vivo, and could dramatically extend the lifetimes of animals in a murine breast cancer model. The system developed in this work therefore merits further investigation as a potential nanotheranostic platform for cancer treatment. STATEMENT OF SIGNIFICANCE: Conventional cancer chemotherapy is accompanied by unavoidable off-target toxicity. Combination therapies, which can ameliorate these issues, are attracting significant attention. Here, the anticancer drug doxorubicin (DOX) was encapsulated in the central cavity of chitosan (CS)-modified hollow mesoporous silica nanoparticles (HMSNs). The prepared system can target drug release to the tumor microenvironment. When exposed to near infrared laser (NIR) irradiation, CuS nanodots located at the surface pores of the HMSNs generate energy, accelerating drug release. In addition, a systematic in vitro and in vivo evaluation confirmed the HMSNs-CS-DOX@CuS platform to give highly effective synergistic chemotherapeutic-photothermal therapy and have effective thermal/photoacoustic dual-imaging properties. This work may open up a new avenue for NIR-enhanced synergistic therapy with simultaneous thermal/photoacoustic dual imaging.
Collapse
Affiliation(s)
- Shiwei Niu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, P.R. China
| | - Xuejing Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jianrong Wu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Feng Gao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, P.R. China
| | - Zi Fu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Xia Chen
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Sheng Lu
- Yunnan Key Laboratory of Digital Orthopaedics, Department of Orthopaedics, the First People's Hospital of Yunnan Province, Kunming 650500, P.R. China.
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China.
| |
Collapse
|
64
|
Mazumdar S, Chitkara D, Mittal A. Exploration and insights into the cellular internalization and intracellular fate of amphiphilic polymeric nanocarriers. Acta Pharm Sin B 2021; 11:903-924. [PMID: 33996406 PMCID: PMC8105776 DOI: 10.1016/j.apsb.2021.02.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/20/2020] [Accepted: 01/18/2021] [Indexed: 01/01/2023] Open
Abstract
The beneficial or deleterious effects of nanomedicines emerge from their complex interactions with intracellular pathways and their subcellular fate. Moreover, the dynamic nature of plasma membrane accounts for the movement of these nanocarriers within the cell towards different organelles thereby not only influencing their pharmacokinetic and pharmacodynamic properties but also bioavailability, therapeutic efficacy and toxicity. Therefore, an in-depth understanding of underlying parameters controlling nanocarrier endocytosis and intracellular fate is essential. In order to direct nanoparticles towards specific sub-cellular organelles the physicochemical attributes of nanocarriers can be manipulated. These include particle size, shape and surface charge/chemistry. Restricting the particle size of nanocarriers below 200 nm contributes to internalization via clathrin and caveolae mediated pathways. Similarly, a moderate negative surface potential confers endolysosomal escape and targeting towards mitochondria, endoplasmic reticulum (ER) and Golgi. This review aims to provide an insight into these physicochemical attributes of nanocarriers fabricated using amphiphilic graft copolymers affecting cellular internalization. Fundamental principles understood from experimental studies have been extrapolated to draw a general conclusion for the designing of optimized nanoparticulate drug delivery systems and enhanced intracellular uptake via specific endocytic pathway.
Collapse
Key Words
- AR, aspect ratio
- Amphiphilic
- CCP, clathrin coated pits
- Cav-1, caveolin-1
- Copolymer
- Cy, cyanine
- DOX, doxorubicin
- ER, endoplasmic reticulum
- FITC, fluorescein isothiocyanate
- HER-2, human epidermal growth factor receptor 2
- IL-2, interleukin
- Internalization
- Intracellular fate
- Nanoparticles
- RBITC, rhodamine B isothiocyanate
- RES, reticuloendothelial system
- Rmax, minimum size threshold value
- Rmin, maximum size threshold value
- SEM, scanning electron microscopy
- SR & LR, short rod and long rod
- TEM, transmission electron microscopy
- mPEG, methoxy poly(ethylene glycol)
Collapse
Affiliation(s)
- Samrat Mazumdar
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS-PILANI), Pilani, Rajasthan 333031, India
| |
Collapse
|
65
|
Rajagopal P, Jayandharan GR, Krishnan UM. Evaluation of the Anticancer Activity of pH-Sensitive Polyketal Nanoparticles for Acute Myeloid Leukemia. Mol Pharm 2021; 18:2015-2031. [PMID: 33780253 DOI: 10.1021/acs.molpharmaceut.0c01243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polyketals are a class of acid-responsive polymers that have been relatively less explored for drug delivery applications compared to polyesters. The degradation of these polymers is accelerated in an acidic medium and does not result in acidic byproducts. Their biocompatibility depends on the diol used for the synthesis. The present work aims to synthesize, characterize, and fabricate nanospheres of an aliphatic polyketal for delivery of the nucleotide analogue cytarabine toward the treatment of acute myeloid leukemia (AML). The internalization mechanism of the nanospheres was probed, and its implication on the nuclear localization and escape from the endo-lysosomal compartments were studied. The drug-loaded polyketal nanoparticles reduced the cell viability to a greater extent compared with the free drug. The effect of the drug-loaded polyketal nanoparticles on the differential gene expression of leukemic cells was investigated for the first time to understand their therapeutic implications. It was found that treatment with drug-loaded polyketal nanoparticles downregulated AML-specific genes involved in cell proliferation and recurrence compared to the free drug. The protein expression studies were performed for selected genes obtained from gene expression analysis. Biodistribution studies showed that the poly(cyclohexane-1,4-diyl acetone dimethylene ketal) (PCADK) nanoparticles exhibit prolonged circulation time. Overall, our results suggest that polyketal-based delivery of cytarabine represents a more effective alternative strategy for AML therapy.
Collapse
Affiliation(s)
- Pratheppa Rajagopal
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India.,The Mehta Family Centre for Engineering In Medicine, Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India.,School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613401, India
| |
Collapse
|
66
|
López-Muñoz R, Treviño ME, Castellanos F, Morales G, Rodríguez-Fernández O, Saavedra S, Licea-Claverie A, Saade H, Enríquez-Medrano FJ, López RG. Loading of doxorubicin on poly(methyl methacrylate-co-methacrylic acid) nanoparticles and release study. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1107-1124. [PMID: 33691605 DOI: 10.1080/09205063.2021.1900652] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Nanoparticles (NP) of 12.7 nm in diameter of the poly(methyl methacrylate (MMA)-co-methacrylic acid (MAA)) copolymer were prepared. 13C-NMR results showed a MMA:MAA molar ratio of 0.64:0.36 in the copolymer, which is similar to the poly(MMA-co-MAA) commercially known as the FDA approved Eudragit S100 (0.67:0.33). The NP prepared in this study were loaded at pH 5 with varying amounts (from 0.54 to 6.91%) of doxorubicin (DOX), an antineoplastic drug. 1H-NMR results indicated the electrostatic interactions between the ionized carboxylic groups of the MAA units in the copolymer and the proton of the glycosidic amine in DOX. Measurements by QLS and TEM indicated that the loading destabilizes the NP, and that for increase stability, they aggregate in a reversible way, forming aggregates with a diameter up to 99.5 nm at a DOX load of 6.91%. The analysis of drug release data at pH 7.4 showed that loaded NP with at least 4.38% DOX release the drug very slowly and follows the Higuchi model; the former suggests that they could remain for long periods in the bloodstream to reach and destroy cancer cells.
Collapse
Affiliation(s)
| | | | | | - Graciela Morales
- Centro de Investigación en Química Aplicada, Saltillo, CH, México
| | | | - Santiago Saavedra
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología, Universidad Autónoma de Nuevo León, Nuevo León, México
| | - Angel Licea-Claverie
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Tijuana, BC, Mexico
| | - Hened Saade
- Centro de Investigación en Química Aplicada, Saltillo, CH, México
| | | | | |
Collapse
|
67
|
Sola F, Canonico B, Montanari M, Volpe A, Barattini C, Pellegrino C, Cesarini E, Guescini M, Battistelli M, Ortolani C, Ventola A, Papa S. Uptake and Intracellular Trafficking Studies of Multiple Dye-Doped Core-Shell Silica Nanoparticles in Lymphoid and Myeloid Cells. Nanotechnol Sci Appl 2021; 14:29-48. [PMID: 33727804 PMCID: PMC7954439 DOI: 10.2147/nsa.s290867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/22/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction Since most biologically active macromolecules are natural nanostructures, operating in the same scale of biomolecules gives the great advantage to enhance the interaction with cellular components. Noteworthy efforts in nanotechnology, particularly in biomedical and pharmaceutical fields, have propelled a high number of studies on the biological effects of nanomaterials. Moreover, the determination of specific physicochemical properties of nanomaterials is crucial for the evaluation and design of novel safe and efficient therapeutics and diagnostic tools. In this in vitro study, we report a physicochemical characterisation of fluorescent silica nanoparticles (NPs), interacting with biological models (U937 and PBMC cells), describing the specific triggered biologic response. Methods Flow Cytometric and Confocal analyses are the main method platforms. However TEM, NTA, DLS, and chemical procedures to synthesize NPs were employed. Results NTB700 NPs, employed in this study, are fluorescent core-shell silica nanoparticles, synthesized through a micelle-assisted method, where the fluorescence energy transfer process, known as FRET, occurs at a high efficiency rate. Using flow cytometry and confocal microscopy, we observed that NTB700 NP uptake seemed to be a rapid, concentration-, energy- and cell type-dependent process, which did not induce significant cytotoxic effects. We did not observe a preferred route of internalization, although their size and the possible aggregated state could influence their extrusion. At this level of analysis, our investigation focuses on lysosome and mitochondria pathways, highlighting that both are involved in NP co-localization. Despite the main mitochondria localization, NPs did not induce a significant increase of intracellular ROS, known inductors of apoptosis, during the time course of analyses. Finally, both lymphoid and myeloid cells are able to release NPs, essential to their biosafety. Discussion These data allow to consider NTB700 NPs a promising platform for future development of a multifunctional system, by combining imaging and localized therapeutic applications in a unique tool.
Collapse
Affiliation(s)
- Federica Sola
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy.,AcZon Srl, Monte San Pietro, BO, 40050, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | | | - Chiara Barattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy.,AcZon Srl, Monte San Pietro, BO, 40050, Italy
| | | | - Erica Cesarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Claudio Ortolani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | | | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| |
Collapse
|
68
|
Xu PY, Zheng X, Kankala RK, Wang SB, Chen AZ. Advances in Indocyanine Green-Based Codelivery Nanoplatforms for Combinatorial Therapy. ACS Biomater Sci Eng 2021; 7:939-962. [PMID: 33539071 DOI: 10.1021/acsbiomaterials.0c01644] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Indocyanine green (ICG), a near-infrared (NIR) agent with an excellent imaging performance, has captivated enormous interest from researchers owing to its excellent therapeutic and imaging abilities. Although various nanoplatforms-based drug delivery systems (DDS) with the ability to overcome the clinical limitations of ICG has been reported, ICG-medicated conventional cancer diagnosis and photorelated therapies still lack in exhibiting the therapeutic efficacy, resulting in incomplete or partly tumor elimination. In the view of addressing these concerns, various DDSs have been engineered for the efficient codelivery of combined therapeutic agents with ICG, aiming to achieve promising therapeutic results due to multifunctional imaging-guided synergistic antitumor effects. In this article, we will systematically review currently available nanoplatforms based on polymers, inorganic, proteins, and metal-organic frameworks (MOFs), among others, for codelivery of ICG along with other therapeutic agents, providing a foundation for future clinical development of ICG. In addition, codelivery systems for ICG and different mechanism-based therapeutic agents will be illustrated. In summary, we conclude the review with the challenges and perspectives of ICG-based versatile nanoplatforms in detail.
Collapse
Affiliation(s)
- Pei-Yao Xu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Xiang Zheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China.,Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| |
Collapse
|
69
|
Andraos C, Gulumian M. Intracellular and extracellular targets as mechanisms of cancer therapy by nanomaterials in relation to their physicochemical properties. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1680. [PMID: 33111484 PMCID: PMC7988657 DOI: 10.1002/wnan.1680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022]
Abstract
Cancer nanomedicine has evolved in recent years and is only expected to increase due to the ease with which nanomaterials (NMs) may be manipulated to the advantage of the cancer patient. The success of nanomedicine is dependent on the cell death mechanism, which in turn is dependent on the organelle initially targeted. The success of cancer nanomedicine is also dependent on other cellular mechanisms such as the induction of autophagy dysfunction, manipulation of the tumor microenvironment (TME) and secretome or induction of host immune responses. Current cancer phototherapies for example, photothermal- or photodynamic therapies as well as radio enhancement also form a major part of cancer nanomedicine. In general, cancer nanomedicine may be grouped into those NMs exhibiting inherent anti-cancer properties that is, self-therapeutic NMs (Group 1), NMs leading to localization of phototherapies or radio-enhancement (Group 2), and NMs as nanocarriers in the absence or presence of external radiation (Group 3). The recent advances of these three groups, together with their advantages and disadvantages as well as their cellular mechanisms and ultimate outcomes are summarized in this review. By exploiting these different intracellular mechanisms involved in initiating cell death pathways, it is possible to synthesize NMs that may have the desirable characteristics to maximize their efficacy in cancer therapy. Therefore, a summary of these important physicochemical characteristics is also presented that need to be considered for optimal cancer cell targeting and initiation of mechanisms that will lead to cancerous cell death. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Charlene Andraos
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
| | - Mary Gulumian
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
- Haematology and Molecular Medicine DepartmentUniversity of the WitwatersrandJohannesburgSouth Africa
- Water Research Group, Unit for Environmental Sciences and ManagementNorth West UniversityPotchefstroomSouth Africa
| |
Collapse
|
70
|
Therapeutic Efficacy and Biodistribution of Paclitaxel-Bound Amphiphilic Cyclodextrin Nanoparticles: Analyses in 3D Tumor Culture and Tumor-Bearing Animals In Vivo. NANOMATERIALS 2021; 11:nano11020515. [PMID: 33670527 PMCID: PMC7922126 DOI: 10.3390/nano11020515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
The uniqueness of paclitaxel’s antimitotic action mechanism has fueled research toward its application in more effective and safer cancer treatments. However, the low water solubility, recrystallization, and side effects hinder the clinical success of classic paclitaxel chemotherapy. The aim of this study was to evaluate the in vivo efficacy and biodistribution of paclitaxel encapsulated in injectable amphiphilic cyclodextrin nanoparticles of different surface charges. It was found that paclitaxel-loaded amphiphilic cyclodextrin nanoparticles showed an antitumoral effect earlier than the drug solution. Moreover, the blank nanoparticles reduced the tumor growth with a similar trend to the paclitaxel solution. At 24 h, the nanoparticles had not accumulated in the heart and lungs according to the biodistribution assessed by in vivo imaging. Therefore, our results indicated that the amphiphilic cyclodextrin nanoparticles are potentially devoid of cardiac toxicity, which limits the clinical use and commercialization of certain polymeric nanoparticles. In conclusion, the amphiphilic cyclodextrin nanoparticles with different surface charge increased the efficiency of paclitaxel in vitro and in vivo. Cyclodextrin nanoparticles could be a good candidate vehicle for intravenous paclitaxel delivery.
Collapse
|
71
|
Fumoto S, Yamamoto T, Okami K, Maemura Y, Terada C, Yamayoshi A, Nishida K. Understanding In Vivo Fate of Nucleic Acid and Gene Medicines for the Rational Design of Drugs. Pharmaceutics 2021; 13:159. [PMID: 33530309 PMCID: PMC7911509 DOI: 10.3390/pharmaceutics13020159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid and genetic medicines are increasingly being developed, owing to their potential to treat a variety of intractable diseases. A comprehensive understanding of the in vivo fate of these agents is vital for the rational design, discovery, and fast and straightforward development of the drugs. In case of intravascular administration of nucleic acids and genetic medicines, interaction with blood components, especially plasma proteins, is unavoidable. However, on the flip side, such interaction can be utilized wisely to manipulate the pharmacokinetics of the agents. In other words, plasma protein binding can help in suppressing the elimination of nucleic acids from the blood stream and deliver naked oligonucleotides and gene carriers into target cells. To control the distribution of these agents in the body, the ligand conjugation method is widely applied. It is also important to understand intracellular localization. In this context, endocytosis pathway, endosomal escape, and nuclear transport should be considered and discussed. Encapsulated nucleic acids and genes must be dissociated from the carriers to exert their activity. In this review, we summarize the in vivo fate of nucleic acid and gene medicines and provide guidelines for the rational design of drugs.
Collapse
Affiliation(s)
- Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; (T.Y.); (K.O.); (Y.M.); (C.T.); (A.Y.); (K.N.)
| | | | | | | | | | | | | |
Collapse
|
72
|
Doroudian M, O' Neill A, Mac Loughlin R, Prina-Mello A, Volkov Y, Donnelly SC. Nanotechnology in pulmonary medicine. Curr Opin Pharmacol 2020; 56:85-92. [PMID: 33341460 PMCID: PMC7746087 DOI: 10.1016/j.coph.2020.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022]
Abstract
Nanotechnology in medicine—nanomedicine—is extensively employed to diagnose, treat, and prevent pulmonary diseases. Over the last few years, this brave new world has made remarkable progress, offering opportunities to address historical clinical challenges in pulmonary diseases including multidrug resistance, adverse side effects of conventional therapeutic agents, novel imaging, and earlier disease detection. Nanomedicine is also being applied to tackle the new emerging infectious diseases, including severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East Respiratory Syndrome Coronavirus (MERS-CoV), influenza A virus subtype H1N1 (A/H1N1), and more recently, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review we provide both a historical overview of the application of nanomedicine to respiratory diseases and more recent cutting-edge approaches such as nanoparticle-mediated combination therapies, novel double-targeted nondrug delivery system for targeting, stimuli-responsive nanoparticles, and theranostic imaging in the diagnosis and treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Mohammad Doroudian
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Ireland; Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Andrew O' Neill
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Ireland
| | - Ronan Mac Loughlin
- Aerogen, IDA Business Park, Dangan, Galway, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Department of Medicine, Trinity College Dublin, Ireland; Nanomedicine Group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Ireland; CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland
| | - Yuri Volkov
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Department of Medicine, Trinity College Dublin, Ireland; Nanomedicine Group, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Ireland; CRANN Institute and AMBER Centre, Trinity College Dublin, Ireland; Department of Histology, Cytology and Embryology, First Moscow State Sechenov Medical University, Moscow, Russian Federation
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Ireland.
| |
Collapse
|
73
|
Docetaxel-loaded block copolymer micelles labeled with 188Re for combined radiochemotherapy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
74
|
Lin CW, Lin SX, Kankala RK, Busa P, Deng JP, Lue SI, Liu CL, Weng CF, Lee CH. Surface-functionalized layered double hydroxide nanocontainers as bile acid sequestrants for lowering hyperlipidemia. Int J Pharm 2020; 590:119921. [PMID: 33027632 DOI: 10.1016/j.ijpharm.2020.119921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 01/01/2023]
Abstract
The surface modification of two-dimensional (2D) nanocontainers with versatile chemical functionalities offers enormous advantages in medicine owing to their altered physicochemical properties. In this study, we demonstrate the fabrication of surface-functionalized layered double hydroxides (LDHs) towards their use as effective intestinal bile acid sequestrants. To demonstrate these aspects, the LDHs are initially modified with an amino silane, N1-(3-trimethoxysilylpropyl) diethylenetriamine (LDHs-N3),which, on the one hand, subsequently used for the fabrication of the dendrimer by repetitive immobilization of ethylene diamine using methyl acrylate as a spacer. On the other hand, these surface-functionalized LDHs are wrapped with an anionic enteric co-polymer to not only prevent the degradation but also increase the stability of these 2D nanoplates in an acidic environment of the stomach to explore the in vivo efficacy. In vitro cholic acid adsorption results showed that these surface-functionalized LDHs displayed tremendous adsorption ability of bile salt. Consequently, the bile salt adsorption results in vivo in mice confirmed that the enteric polymer-coated diethylenetriamine silane-modified LDHs, resulting in the reduced cholesterol by 8.2% in the high fat diet-fed mice compared to that of the oil treatment group with augmented 28% of cholesterol, which gained weight by 6.7% in 4 weeks. Notably, the relative organ (liver and kidney) weight analysis and the tissue section of histology results indicated that the modified LDHs showed high biocompatibility in vivo. Together, our findings validate that these surface-functionalized 2D nanoplates have great potential as effective intestinal bile acid sequestrants.
Collapse
Affiliation(s)
- Chiao-Wen Lin
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Shi-Xiang Lin
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Ranjith Kumar Kankala
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan; College of Chemical Engineering, Huaqiao University, Xiamen 361021, PR China
| | - Prabhakar Busa
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Jin-Pei Deng
- Department of Chemistry, Tamkang University, New Taipei City 251, Taiwan
| | - Sheng-I Lue
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan; Department of Physiology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chen-Lun Liu
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Ching-Feng Weng
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan
| | - Chia-Hung Lee
- Department of Life Science, National Dong Hwa University, Hualien 97401, Taiwan.
| |
Collapse
|
75
|
Wang X, Qiu Y, Wang M, Zhang C, Zhang T, Zhou H, Zhao W, Zhao W, Xia G, Shao R. Endocytosis and Organelle Targeting of Nanomedicines in Cancer Therapy. Int J Nanomedicine 2020; 15:9447-9467. [PMID: 33268987 PMCID: PMC7701161 DOI: 10.2147/ijn.s274289] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines (NMs) have played an increasing role in cancer therapy as carriers to efficiently deliver therapeutics into tumor cells. For this application, the uptake of NMs by tumor cells is usually a prerequisite to deliver the cargo to intracellular locations, which mainly relies on endocytosis. NMs can enter cells through a variety of endocytosis pathways. Different endocytosis pathways exhibit different intracellular trafficking routes and diverse subcellular localizations. Therefore, a comprehensive understanding of endocytosis mechanisms is necessary for increasing cellular entry efficiency and to trace the fate of NMs after internalization. This review focuses on endocytosis pathways of NMs in tumor cells, mainly including clathrin- and caveolae-mediated endocytosis pathways, involving effector molecules, expression difference of those molecules between normal and tumor cells, as well as the intracellular trafficking route of corresponding endocytosis vesicles. Then, the latest strategies for NMs to actively employ endocytosis are described, including improving tumor cellular uptake of NMs by receptor-mediated endocytosis, transporter-mediated endocytosis and enabling drug activity by changing intracellular routes. Finally, active targeting strategies towards intracellular organelles are also mentioned. This review will be helpful not only in explicating endocytosis and the trafficking process of NMs and elucidating anti-tumor mechanisms inside the cell but also in rendering new ideas for the design of highly efficacious and cancer-targeted NMs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mengyan Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Conghui Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tianshu Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Huimin Zhou
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenxia Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rongguang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
76
|
Kankala RK, Wang SB, Chen AZ. Nanoarchitecting Hierarchical Mesoporous Siliceous Frameworks: A New Way Forward. iScience 2020; 23:101687. [PMID: 33163941 PMCID: PMC7607446 DOI: 10.1016/j.isci.2020.101687] [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] [Indexed: 01/29/2023] Open
Abstract
Owing to their attractive physicochemical and morphological attributes, mesoporous silica nanoparticles (MSNs) have attracted increasing attention over the past two decades for their utilization in diversified fields. Despite the success, these highly stable siliceous frameworks often suffer from several shortcomings of compatibility issues, uncontrollable degradability leading to long-term retention in vivo, and substantial unpredictable toxicity risks, as well as deprived drug encapsulation efficiency, which could limit their applicability in medicine. Along this line, various advancements have been made in re-engineering the stable siliceous frameworks, such as the incorporation of diverse molecular organic, as well as inorganic (cationic and anionic) species and monitoring the processing, as well as formulation parameters, resulting in the hetero-nanostructures of irregular-shaped (Janus and multi-podal) and dynamically-modulated (deformable solids) architectures with high morphological complexity. Insightfully, this review gives a brief emphasis on re-engineering such stable siliceous frameworks through modifying their intrinsic structural and physicochemical attributes. In conclusion, we recapitulate the review with exciting perspectives.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Shi-Bin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| | - Ai-Zheng Chen
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian 361021, P. R. China
| |
Collapse
|
77
|
Homsirikamol C, Suvanasuthi S, Viravaidya-Pasuwat K. Inclusion of IR-820 into Soybean-Phosphatides-Based Nanoparticles for Near-Infrared-Triggered Release and Endolysosomal Escape in HaCaT Keratinocytes at Insignificant Cytotoxic Level. Int J Nanomedicine 2020; 15:8717-8737. [PMID: 33192063 PMCID: PMC7654534 DOI: 10.2147/ijn.s267119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/22/2020] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The degradation of drugs within endolysosomes has been widely addressed as a cause of poor bioavailability. One of the strategies to allow molecules to escape from a destructive fate is to introduce a photosensitizing moiety into a drug carrier enabling the permeabilization of endosomes and endolysosomes upon irradiation. This paper presents an alternative delivery nanosystem composed of cost-effective soybean phosphatides mixed with IR-820, a near-infrared (NIR) sensitizer, to load various active compounds and trigger an endolysosomal escape with a low cytotoxic effect. METHODS IR-820-incorporated phosphatides-based nanoparticles were formulated using a thin-film hydration method to encapsulate different molecular probes and a drug model. The nanoparticles were characterized in vitro using dynamic light scattering, transmission electron microscopy, as well as ultraviolet-visible and fluorescence spectroscopy techniques. The NIR-corresponding generation of the photochemical products, the content release, and the cytotoxicity toward the HaCaT keratinocyte cell line were evaluated. The cellular internalization and endolysosomal escape were monitored using a cytochemical marker and fluorescent probes with a colocalization analysis. RESULTS The IR-820-combined nanoparticles revealed the NIR-triggered changes in the singlet oxygen presence, nanoparticle architecture, and release rate without being cytotoxic. Additionally, the nanoplatform appeared to enhance cellular uptake of the macromolecules. The localization of the cytochemical marker and the colocalization analysis on the fluorescence signals of the encapsulated fluorophore and the lysosome-labeling reporter implied the transient endolysosomal escape of the cargo within the HaCaT cells after NIR irradiation. CONCLUSION The inclusion of IR-820 into a soybean-phosphatides base ingredient provides NIR responsiveness, particularly the endolysosomal escape of the payload, to the formulated nanoparticles, while preserving the beneficial properties as a drug carrier. This alternative delivery nanomedicine system has future potential to provide high bioavailability of cytosolic drugs utilizing time- and spatial-controllable NIR triggerability as well as the synergistic therapeutic effects with NIR-biomodulation.
Collapse
Affiliation(s)
- Chaiyarerk Homsirikamol
- Mammalian Cell Culture Laboratory, Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Saroj Suvanasuthi
- Hair Diseases and Hair Transplantation Division, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Mammalian Cell Culture Laboratory, Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- Department of Chemical Engineering, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
78
|
Fu X, Shi Y, Qi T, Qiu S, Huang Y, Zhao X, Sun Q, Lin G. Precise design strategies of nanomedicine for improving cancer therapeutic efficacy using subcellular targeting. Signal Transduct Target Ther 2020; 5:262. [PMID: 33154350 PMCID: PMC7644763 DOI: 10.1038/s41392-020-00342-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Therapeutic efficacy against cancer relies heavily on the ability of the therapeutic agents to reach their final targets. The optimal targets of most cancer therapeutic agents are usually biological macromolecules at the subcellular level, which play a key role in carcinogenesis. Therefore, to improve the therapeutic efficiency of drugs, researchers need to focus on delivering not only the therapeutic agents to the target tissues and cells but also the drugs to the relevant subcellular structures. In this review, we discuss the most recent construction strategies and release patterns of various cancer cell subcellular-targeting nanoformulations, aiming at providing guidance in the overall design of precise nanomedicine. Additionally, future challenges and potential perspectives are illustrated in the hope of enhancing anticancer efficacy and accelerating the translational progress of precise nanomedicine.
Collapse
Affiliation(s)
- Xianglei Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yanbin Shi
- School of Mechanical and Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, Shandong, China
| | - Tongtong Qi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Shengnan Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yi Huang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaogang Zhao
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Qifeng Sun
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Guimei Lin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
79
|
Liu CG, Tang HX, Zheng X, Yang DY, Zhang Y, Zhang JT, Kankala RK, Wang SB, Liu G, Chen AZ. Near-Infrared-Activated Lysosome Pathway Death Induced by ROS Generated from Layered Double Hydroxide-Copper Sulfide Nanocomposites. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40673-40683. [PMID: 32786245 DOI: 10.1021/acsami.0c11739] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The overdeveloped lysosomes in cancer cells are gaining increasing attention toward more precise and effective organelle-targeted cancer therapy. It is suggested that rod/plate-like nanomaterials with an appropriate size exhibited a greater quantity and longer-term lysosomal enrichment, as the shape plays a notable role in the nanomaterial transmembrane process and subcellular behaviors. Herein, a biodegradable platform based on layered double hydroxide-copper sulfide nanocomposites (LDH-CuS NCs) is successfully prepared via in situ growth of CuS nanodots on LDH nanoplates. The as-prepared LDH-CuS NCs exhibited not only high photothermal conversion and near-infrared (NIR)-induced chemodynamic and photodynamic therapeutic efficacies, but also could achieve real-time in vivo photoacoustic imaging (PAI) of the entire tumor. LDH-CuS NCs accumulated in lysosomes would then generate extensive subcellular reactive oxygen species (ROS) in situ, leading to lysosomal membrane permeabilization (LMP) pathway-associated cell death both in vitro and in vivo.
Collapse
Affiliation(s)
- Chen-Guang Liu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Han-Xiao Tang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Xiang Zheng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Da-Yun Yang
- Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, P. R. China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Jian-Ting Zhang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, P. R. China
| |
Collapse
|
80
|
Hamida RS, Ali MA, Redhwan A, Bin-Meferij MM. Cyanobacteria - A Promising Platform in Green Nanotechnology: A Review on Nanoparticles Fabrication and Their Prospective Applications. Int J Nanomedicine 2020; 15:6033-6066. [PMID: 32884261 PMCID: PMC7434529 DOI: 10.2147/ijn.s256134] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Green synthesis of nanoparticles (NPs) is a global ecofriendly method to develop and produce nanomaterials with unique biological, physical, and chemical properties. Recently, attention has shifted toward biological synthesis, owing to the disadvantages of physical and chemical synthesis, which include toxic yields, time and energy consumption, and high cost. Many natural sources are used in green fabrication processes, including yeasts, plants, fungi, actinomycetes, algae, and cyanobacteria. Cyanobacteria are among the most beneficial natural candidates used in the biosynthesis of NPs, due to their ability to accumulate heavy metals from their environment. They also contain a variety of bioactive compounds, such as pigments and enzymes, that may act as reducing and stabilizing agents. Cyanobacteria-mediated NPs have potential antibacterial, antifungal, antialgal, anticancer, and photocatalytic activities. The present review paper highlights the characteristics and applications in various fields of NPs produced by cyanobacteria-mediated synthesis.
Collapse
Affiliation(s)
- Reham Samir Hamida
- Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Abdelaal Ali
- Biotechnology Unit, Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Alya Redhwan
- Department of Health, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | |
Collapse
|
81
|
Zhao Y, Chen BQ, Kankala RK, Wang SB, Chen AZ. Recent Advances in Combination of Copper Chalcogenide-Based Photothermal and Reactive Oxygen Species-Related Therapies. ACS Biomater Sci Eng 2020; 6:4799-4815. [DOI: 10.1021/acsbiomaterials.0c00830] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yi Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Biao-Qi Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| |
Collapse
|
82
|
Supercritical antisolvent process-assisted fabrication of chrysin-polyvinylpyrrolidone sub-microparticles for improved anticancer efficiency. J Supercrit Fluids 2020. [DOI: 10.1016/j.supflu.2020.104847] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
83
|
Hamida RS, Albasher G, Bin-Meferij MM. Oxidative Stress and Apoptotic Responses Elicited by Nostoc-Synthesized Silver Nanoparticles against Different Cancer Cell Lines. Cancers (Basel) 2020; 12:E2099. [PMID: 32731591 PMCID: PMC7464693 DOI: 10.3390/cancers12082099] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Green nanoparticles represent a revolution in bionanotechnology, providing opportunities to fight life-threatening diseases, such as cancer, with less risk to the environment and to human health. Here, for the first time, we systematically investigated the anticancer activity and possible mechanism of novel silver nanoparticles (N-SNPs) synthesized by Nostoc Bahar M against the MCF-7 breast cancer cells, HCT-116 colorectal adenocarcinoma cells, and HepG2 liver cancer cells, using cell viability assays, morphological characterization with inverted light and transmission electron microscopy, antioxidants and enzymes (glutathione peroxidase (GPx), glutathione (GSH), adenosine triphosphatase (ATPase), and lactate dehydrogenase (LDH)), and western blotting (protein kinase B (Akt), phosphorylated-Akt (p-Akt), mammalian target of rapamycin (mTOR), B-cell lymphoma 2 (Bcl-2), tumor suppressor (p53), and caspase 3). N-SNPs decreased the viability of MCF-7, HCT-116, and HepG2 cells, with half-maximal inhibitory concentrations of 54, 56, and 80 µg/mL, respectively. They also significantly increased LDH leakage, enhanced oxidative stress via effects on antioxidative markers, and caused metabolic stress by significantly decreasing ATPase levels. N-SNPs caused extensive ultrastructural alterations in cell and nuclear structures, as well as in various organelles. Furthermore, N-SNPs triggered apoptosis via the activation of caspase 3 and p53, and suppressed the mTOR signaling pathway via downregulating apoptosis-evading proteins in MCF-7, HCT-116, and HepG2 cells. Ultrastructural analysis, together with biochemical and molecular analyses, revealed that N-SNPs enhanced apoptosis via the induction of oxidative stress and/or through direct interactions with cellular structures in all tested cells. The cytotoxicity of Nostoc-mediated SNPs represents a new strategy for cancer treatment via targeting various cell death pathways. However, the potential of N-SNPs to be usable and biocompatible anticancer drug will depend on their toxicity against normal cells.
Collapse
Affiliation(s)
- Reham Samir Hamida
- Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21500, Egypt
| | - Gadah Albasher
- Zoology Department, College of Science, King Saud University, Riyadh 11543, Saudi Arabia;
| | - Mashael Mohammed Bin-Meferij
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11543, Saudi Arabia
| |
Collapse
|
84
|
Exploiting Fruit Waste Grape Pomace for Silver Nanoparticles Synthesis, Assessing Their Antioxidant, Antidiabetic Potential and Antibacterial Activity Against Human Pathogens: A Novel Approach. NANOMATERIALS 2020; 10:nano10081457. [PMID: 32722404 PMCID: PMC7466627 DOI: 10.3390/nano10081457] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
Abstract
Grape pomace, a most abundant and renewable wine industry waste product was utilized as a suitable reducing, capping, and stabilizing biomolecules for green synthesis of silver nanoparticles (AgNPs). The physicochemical properties of biosynthesized grape pomace extract (GPE)-AgNPs were duly appraised via UV-Visible spectroscopy, X-ray diffractometer (XRD), Fourier-transform infrared spectroscopy (FTIR), and transmission electron microscopy. The analytical studies revealed that the GPE-AgNPs were well formed and stable in nature. The functional groups of organic molecules of GPE are present on the surface of AgNPs with average NPs diameter in the range of 20-35 nm. GPE-AgNPs exhibited significant free radical scavenging activity mainly DPPH radical (IC50, 50.0 ± 2.25 μg/mL) and ABTS radical (IC50, 38.46 ± 1.14 μg/mL). Additionally, the synthesized AgNPs showed noticeable inhibition of carbohydrate hydrolyzing enzymes mainly, α-amylase (IC50, 60.2 ± 2.15 μg/mL) and α-glucosidase (IC50, 62.5 ± 2.75 μg/mL). The GPE fabricated AgNPs showed noteworthy antibacterial potential against infectious bacteria viz., Escherichia coli and Staphylococcus aureus. The reaction mechanism of antibacterial activity was studied by measuring the bacterial cell membrane breakage and cytoplasmic contents, mainly, nucleic acid, proteins, and reducing sugar. Therefore, this research attempt illustrated the potential of GPE as a novel source intended for the biosynthesis of AgNPs that may open up new horizons in the field of nanomedicine.
Collapse
|
85
|
Tang HX, Cai YY, Liu CG, Zhang JT, Kankala RK, Wang SB, Chen AZ. Sub-micronization of disulfiram and disulfiram-copper complexes by Rapid expansion of supercritical solution toward augmented anticancer effect. J CO2 UTIL 2020. [DOI: 10.1016/j.jcou.2020.101187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
86
|
Xu PY, Fu CP, Kankala RK, Wang SB, Chen AZ. Supercritical carbon dioxide-assisted nanonization of dihydromyricetin for anticancer and bacterial biofilm inhibition efficacies. J Supercrit Fluids 2020. [DOI: 10.1016/j.supflu.2020.104840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
87
|
Kumar A, Ahmad A, Vyawahare A, Khan R. Membrane Trafficking and Subcellular Drug Targeting Pathways. Front Pharmacol 2020; 11:629. [PMID: 32536862 PMCID: PMC7267071 DOI: 10.3389/fphar.2020.00629] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/21/2020] [Indexed: 12/29/2022] Open
Abstract
The movement of micro and macro molecules into and within a cell significantly governs several of their pharmacokinetic and pharmacodynamic parameters, thus regulating the cellular response to exogenous and endogenous stimuli. Trafficking of various pharmacological agents and other bioactive molecules throughout and within the cell is necessary for the fidelity of the cells but has been poorly investigated. Novel strategies against cancer and microbial infections need a deeper understanding of membrane as well as subcellular trafficking pathways and essentially regulate several aspects of the initiation and spread of anti-microbial and anti-cancer drug resistance. Furthermore, in order to avail the maximum possible bioavailability and therapeutic efficacy and to restrict the unwanted toxicity of pharmacological bioactives, these sometimes need to be functionalized with targeting ligands to regulate the subcellular trafficking and to enhance the localization. In the recent past the scenario drug targeting has primarily focused on targeting tissue components and cell vicinities, however, it is the membranous and subcellular trafficking system that directs the molecules to plausible locations. The effectiveness of the delivery platforms largely depends on their physicochemical nature, intracellular barriers, and biodistribution of the drugs, pharmacokinetics and pharmacodynamic paradigms. Most subcellular organelles possess some peculiar characteristics by which membranous and subcellular targeting can be manipulated, such as negative transmembrane potential in mitochondria, intraluminal delta pH in a lysosome, and many others. Many specialized methods, which positively promote the subcellular targeting and restrict the off-targeting of the bioactive molecules, exist. Recent advancements in designing the carrier molecules enable the handling of membrane trafficking to facilitate the delivery of active compounds to subcellular localizations. This review aims to cover membrane trafficking pathways which promote the delivery of the active molecule in to the subcellular locations, the associated pathways of the subcellular drug delivery system, and the role of the carrier system in drug delivery techniques.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Mohali, India
| | - Anas Ahmad
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Mohali, India
| | - Akshay Vyawahare
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Mohali, India
| | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Mohali, India
| |
Collapse
|
88
|
Abstract
Being originally discovered as cellular recycling bins, lysosomes are today recognized as versatile signaling organelles that control a wide range of cellular functions that are essential not only for the well-being of normal cells but also for malignant transformation and cancer progression. In addition to their core functions in waste disposal and recycling of macromolecules and energy, lysosomes serve as an indispensable support system for malignant phenotype by promoting cell growth, cytoprotective autophagy, drug resistance, pH homeostasis, invasion, metastasis, and genomic integrity. On the other hand, malignant transformation reduces the stability of lysosomal membranes rendering cancer cells sensitive to lysosome-dependent cell death. Notably, many clinically approved cationic amphiphilic drugs widely used for the treatment of other diseases accumulate in lysosomes, interfere with their cancer-promoting and cancer-supporting functions and destabilize their membranes thereby opening intriguing possibilities for cancer therapy. Here, we review the emerging evidence that supports the supplementation of current cancer therapies with lysosome-targeting cationic amphiphilic drugs.
Collapse
|