1
|
Yu H, Yu J, Yao G. Recent Advances in Aptamers-Based Nanosystems for Diagnosis and Therapy of Cardiovascular Diseases: An Updated Review. Int J Nanomedicine 2025; 20:2427-2443. [PMID: 40034222 PMCID: PMC11873322 DOI: 10.2147/ijn.s507715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
The increasing global prevalence of cardiovascular diseases highlights the urgent need for innovative diagnostic and therapeutic strategies. Aptamers, small single-stranded nucleic acid molecules with exceptional specificity and affinity for target biomolecules, have emerged as promising tools for precise diagnostics and targeted therapies. Their selective binding capabilities provide valuable insights into the molecular mechanisms underlying cardiovascular conditions. When integrated into nanosystems, aptamers enhance the delivery, bioavailability, and stability of diagnostic and therapeutic agents, addressing challenges of solubility and degradation. This integration enables more targeted drug delivery, advanced imaging techniques, and improved therapeutic interventions, ultimately improving the management of cardiovascular diseases. Recent advancements in aptamer selection methodologies, coupled with their unique three-dimensional structures, have significantly expanded their application potential in cardiovascular health. By combining aptamers with nanosystems, novel approaches to cardiovascular disease diagnosis and treatment are emerging, promising enhanced efficacy, safety, and precision. This review explores recent progress in the development and application of aptamer-based nanosystems in cardiovascular diagnostics and therapies.
Collapse
Affiliation(s)
- Hongqin Yu
- Department of Cardiovascular Medicine, Yantai Mountain Hospital, Yantai, 264000, People’s Republic of China
| | - Jie Yu
- Department of Cardiovascular Medicine, Yantai Mountain Hospital, Yantai, 264000, People’s Republic of China
| | - Gang Yao
- Department of Cardiovascular Medicine, Yantai Mountain Hospital, Yantai, 264000, People’s Republic of China
| |
Collapse
|
2
|
Liu H, Liu S, Ma P, Ma L, Liu Y, Zhao F, Zhou R. Development and Evaluation of Aloperine-Loaded Nanostructured Lipid Carriers for the Treatment of Pulmonary Arterial Hypertension. Int J Nanomedicine 2025; 20:871-886. [PMID: 39867311 PMCID: PMC11761852 DOI: 10.2147/ijn.s489133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Objective This study focuses on the development and evaluation of nanostructured lipid carriers (NLCs) loaded with aloperine as a potential therapeutic approach for the treatment of pulmonary arterial hypertension. Methods The NLCs were designed to enhance the solubility, stability, and bioavailability of aloperine, a compound with vasodilatory and anti-inflammatory properties. Through a series of experiments including single-factor experimentation, transmission electron microscopy, high-performance liquid chromatography, in vivo pharmacokinetics, and tissue distribution studies, we assessed the physicochemical properties, drug release profiles, and in vitro and in vivo performance of this novel nanocarrier. Results The prepared aloperine-loaded NLCs exhibited a milky white and translucent suspension appearance, presenting a quasi-spherical shape under a transmission electron microscope, with an average particle size of (509.48±30.04) nm and an entrapment efficiency of (64.18±1.14)%. The drug release profile demonstrated good sustained-release characteristics in vitro, and the formulation remained stable for up to 15 days when stored at 4°C. Compared to the aloperine solution group, the t1/2, AUC(0→t), AUC(0→∞), MRT(0→t), and clearance rate of the aloperine-loaded NLCs were 2.3, 2.96, 3.06, 3.03, and 0.22 times higher, respectively. This indicates that formulating aloperine into NLCs can prolong its circulation time in the body. Furthermore, the concentrations of aloperine in the lungs of the NLCs group were 1.79, 3.78, and 2.30 times higher than those in the solution group at three time points (0.25 h, 1.5 h, 4 h), suggesting that NLCs can increase the accumulation of aloperine in the lungs. Conclusion Our findings suggest that NLCs loaded with aloperine could offer a promising strategy for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Hui Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Siyun Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Pengsheng Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Long Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Yuxin Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Fang Zhao
- General Hospital of Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Ru Zhou
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
- Ningxia Characteristic Traditional Chinese Medicine Modernization Engineering Technology Research Center, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| |
Collapse
|
3
|
Chen S, Wu Z. Targeting tumor microenvironments with gold nanoparticles for enhanced photothermal therapy. ONCOLOGIE 2024; 26:899-912. [DOI: 10.1515/oncologie-2024-0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Abstract
Gold nano-drug delivery system-mediated photothermal therapy (PTT) has been widely studied in the field of anti-tumor. In order to achieve accurate drug release and improve photothermal efficiency, nano-drug delivery strategies targeting tumor microenvironment (TME) have become a hot research topic in recent years. This paper introduces four characteristics of the TME: hypoxia, low pH, high level of reactive oxygen species (ROS), and overexpression of enzymes. These differences between tumor and normal tissue become effective targets for tumor therapy. This paper summarizes the gold nano-drug delivery system that can target these four characteristics, so as to realize a large amount of drug aggregation at the tumor site and achieve efficient photothermal therapy. Moreover, the multi-response nano-drug delivery system can further control drug delivery and improve therapeutic effects. Finally, this paper also summarizes the gold nanoparticles for tumor therapy that have entered clinical trials so far. The purpose of this review is to discuss the research progress of enhanced photothermal therapy with gold nano-drug delivery systems targeting the TME, with a view to providing a reference for the future development of novel anti-tumor nanoplatforms and the clinical translation of gold nanoparticles.
Collapse
Affiliation(s)
- Sisi Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University , Hangzhou , China
| | - Zhibing Wu
- Department of Oncology , 584020 Affiliated Zhejiang Hospital of Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
4
|
Simmons A, Mihalek O, Bimonte Nelson HA, Sirianni RW, Stabenfeldt SE. Acute brain injury and nanomedicine: sex as a biological variable. FRONTIERS IN BIOMATERIALS SCIENCE 2024; 3:1348165. [PMID: 39450372 PMCID: PMC11500709 DOI: 10.3389/fbiom.2024.1348165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Sex as a biological variable has been recognized for decades to be a critical aspect of the drug development process, as differences in drug pharmacology and toxicity in female versus male subjects can drive the success or failure of new therapeutics. These concepts in development of traditional drug systems have only recently begun to be applied for advancing nanomedicine systems that are designed for drug delivery or imaging in the central nervous system (CNS). This review provides a comprehensive overview of the current state of two fields of research - nanomedicine and acute brain injury-centering on sex as a biological variable. We highlight areas of each field that provide foundational understanding of sex as a biological variable in nanomedicine, brain development, immune response, and pathophysiology of traumatic brain injury and stroke. We describe current knowledge on female versus male physiology as well as a growing number of empirical reports that directly address sex as a biological variable in these contexts. In sum, the data make clear two key observations. First, the manner in which sex affects nanomedicine distribution, toxicity, or efficacy is important, complex, and depends on the specific nanoparticle system under considerations; second, although field knowledge is accumulating to enable us to understand sex as a biological variable in the fields of nanomedicine and acute brain injury, there are critical gaps in knowledge that will need to be addressed. We anticipate that understanding sex as a biological variable in the development of nanomedicine systems to treat acute CNS injury will be an important determinant of their success.
Collapse
Affiliation(s)
- Amberlyn Simmons
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Olivia Mihalek
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | | | - Rachael W. Sirianni
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
5
|
Wen X, Ou L, Cutshaw G, Uthaman S, Ou YC, Zhu T, Szakas S, Carney B, Houghton J, Gundlach-Graham A, Rafat M, Yang K, Bardhan R. Physicochemical Properties and Route of Systemic Delivery Control the In Vivo Dynamics and Breakdown of Radiolabeled Gold Nanostars. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204293. [PMID: 36965074 PMCID: PMC10518372 DOI: 10.1002/smll.202204293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 02/22/2023] [Indexed: 06/18/2023]
Abstract
The in vivo dynamics of nanoparticles requires a mechanistic understanding of multiple factors. Here, for the first time, the surprising breakdown of functionalized gold nanostars (F-AuNSs) conjugated with antibodies and 64 Cu radiolabels in vivo and in artificial lysosomal fluid ex vivo, is shown. The short-term biodistribution of F-AuNSs is driven by the route of systemic delivery (intravenous vs intraperitoneal) and long-term fate is controlled by the tissue type in vivo. In vitro studies including endocytosis pathways, intracellular trafficking, and opsonization, are combined with in vivo studies integrating a milieu of spectroscopy and microcopy techniques that show F-AuNSs dynamics is driven by their physicochemical properties and route of delivery. F-AuNSs break down into sub-20 nm broken nanoparticles as early as 7 days postinjection. Martini coarse-grained simulations are performed to support the in vivo findings. Simulations suggest that shape, size, and charge of the broken nanoparticles, and composition of the lipid membrane depicting various tissues govern the interaction of the nanoparticles with the membrane, and the rate of translocation across the membrane to ultimately enable tissue clearance. The fundamental study addresses critical gaps in the knowledge regarding the fate of nanoparticles in vivo that remain a bottleneck in their clinical translation.
Collapse
Affiliation(s)
- Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
| | - Luping Ou
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Gabriel Cutshaw
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| | - Saji Uthaman
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| | - Yu-Chuan Ou
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Sarah Szakas
- Department of Chemistry, Iowa State University, Ames, IA, 50011, USA
| | - Brandon Carney
- Department of Radiology, Stony Brook University, Stony Brook, New York, NY, 11794, USA
| | - Jacob Houghton
- Department of Radiology, Stony Brook University, Stony Brook, New York, NY, 11794, USA
| | | | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and Technology, Soochow University, Suzhou, 215006, China
| | - Rizia Bardhan
- Nanovaccine Institute, Iowa State University, Ames, IA, 50012, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50012, USA
| |
Collapse
|
6
|
Li K, Liu Y, Lou B, Tan Y, Chen L, Liu Z. DNA-directed assembly of nanomaterials and their biomedical applications. Int J Biol Macromol 2023:125551. [PMID: 37356694 DOI: 10.1016/j.ijbiomac.2023.125551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
In the past decades, DNA has been widely used in the field of nanostructures due to its unique programmable properties. Besides being used to form its own diverse structures such as the assembly of DNA origami, DNA can also be used for the assembly of nanostructures with other materials. In this review, different strategies for the functionalization of DNA on nanoparticle surfaces are listed, and the roles of DNA in the assembly of nanostructures as well as the influencing factors are discussed. Finally, the biomedical applications of DNA-assembled nanostructures were summarized. This review provided new insight into the application of DNA in nanostructure assembly.
Collapse
Affiliation(s)
- Ke Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Beibei Lou
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yifu Tan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Liwei Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan Province, PR China.
| |
Collapse
|
7
|
Googasian JS, Skrabalak SE. Practical Considerations for Simulating the Plasmonic Properties of Metal Nanoparticles. ACS PHYSICAL CHEMISTRY AU 2023; 3:252-262. [PMID: 37249938 PMCID: PMC10214510 DOI: 10.1021/acsphyschemau.2c00064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 05/31/2023]
Abstract
Simulating the plasmonic properties of colloidally derived metal nanoparticles with accuracy to their experimentally observed measurements is challenging due to the many structural and compositional parameters that influence their scattering and absorption properties. Correlation between single nanoparticle scattering measurements and simulated spectra emphasize these strong structural and compositional relationships, providing insight into the design of plasmonic nanoparticles. This Perspective builds from this history to highlight how the structural features of models used in simulation methods such as those based on the Finite-Difference Time-Domain (FDTD) method and Discrete Dipole Approximation (DDA) are of critical consideration for correlation with experiment and ultimately prediction of new nanoparticle properties. High-level characterizations such as electron tomography are discussed as ways to advance the accuracy of models used in such simulations, allowing the plasmonic properties of structurally complex nanoparticles to be better understood. However, we also note that the field is far from bringing experiment and simulation into agreement for plasmonic nanoparticles with complex compositions, reflecting analytical challenges that inhibit accurate model generation. Potential directions for addressing these challenges are also presented.
Collapse
Affiliation(s)
- Jack S. Googasian
- Department of Chemistry, Indiana
University—Bloomington, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Sara E. Skrabalak
- Department of Chemistry, Indiana
University—Bloomington, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
8
|
Xiao Y, Pandey K, Nicolás-Boluda A, Onidas D, Nizard P, Carn F, Lucas T, Gateau J, Martin-Molina A, Quesada-Pérez M, Del Mar Ramos-Tejada M, Gazeau F, Luo Y, Mangeney C. Synergic Thermo- and pH-Sensitive Hybrid Microgels Loaded with Fluorescent Dyes and Ultrasmall Gold Nanoparticles for Photoacoustic Imaging and Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54439-54457. [PMID: 36468426 DOI: 10.1021/acsami.2c12796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Smart microgels (μGels) made of polymeric particles doped with inorganic nanoparticles have emerged recently as promising multifunctional materials for nanomedicine applications. However, the synthesis of these hybrid materials is still a challenging task with the necessity to control several features, such as particle sizes and doping levels, in order to tailor their final properties in relation to the targeted application. We report herein an innovative modular strategy to achieve the rational design of well-defined and densely filled hybrid particles. It is based on the assembly of the different building blocks, i.e., μGels, dyes, and small gold nanoparticles (<4 nm), and the tuning of nanoparticle loading within the polymer matrix through successive incubation steps. The characterization of the final hybrid networks using UV-vis absorption, fluorescence, transmission electron microscopy, dynamic light scattering, and small-angle X-ray scattering revealed that they uniquely combine the properties of hydrogel particles, including high loading capacity and stimuli-responsive behavior, the photoluminescent properties of dyes (rhodamine 6G, methylene blue and cyanine 7.5), and the features of gold nanoparticle assembly. Interestingly, in response to pH and temperature stimuli, the smart hybrid μGels can shrink, leading to the aggregation of the gold nanoparticles trapped inside the polymer matrix. This stimuli-responsive behavior results in plasmon band broadening and red shift toward the near-infrared region (NIR), opening promising prospects in biomedical science. Particularly, the potential of these smart hybrid nanoplatforms for photoactivated hyperthermia, photoacoustic imaging, cellular internalization, intracellular imaging, and photothermal therapy was assessed, demonstrating well controlled multimodal opportunities for theranostics.
Collapse
Affiliation(s)
- Yu Xiao
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| | - Kartikey Pandey
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| | - Alba Nicolás-Boluda
- CNRS Matière et Systèmes Complexes MSC, Université Paris Cité, ParisF-75006, France
| | - Delphine Onidas
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| | - Philippe Nizard
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| | - Florent Carn
- CNRS Matière et Systèmes Complexes MSC, Université Paris Cité, ParisF-75006, France
| | - Théotim Lucas
- CNRS Matière et Systèmes Complexes MSC, Université Paris Cité, ParisF-75006, France
- CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB, Sorbonne Université, ParisF-75006, France
| | - Jérôme Gateau
- CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB, Sorbonne Université, ParisF-75006, France
| | - Alberto Martin-Molina
- Departamento de Física Aplicada, Universidad de Granada, Campus de Fuentenueva s/n, Granada18071, Spain
- Instituto Carlos I de Física Teórica y Computacional, Universidad de Granada, Campus de Fuentenueva s/n, Granada18071, Spain
| | - Manuel Quesada-Pérez
- Departamento de Física, Escuela Politécnica Superior de Linares, Universidad de Jaén, Linares, Jaén23700, Spain
| | - Maria Del Mar Ramos-Tejada
- Departamento de Física, Escuela Politécnica Superior de Linares, Universidad de Jaén, Linares, Jaén23700, Spain
| | - Florence Gazeau
- CNRS Matière et Systèmes Complexes MSC, Université Paris Cité, ParisF-75006, France
| | - Yun Luo
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| | - Claire Mangeney
- CNRS Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, ParisF-75006, France
| |
Collapse
|
9
|
Unida V, Vindigni G, Raniolo S, Stolfi C, Desideri A, Biocca S. Folate-Functionalization Enhances Cytotoxicity of Multivalent DNA Nanocages on Triple-Negative Breast Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14122610. [PMID: 36559104 PMCID: PMC9786333 DOI: 10.3390/pharmaceutics14122610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
DNA is an excellent programmable polymer for the generation of self-assembled multivalent nanostructures useful for biomedical applications. Herein, we developed (i) folate-functionalized nanocages (Fol-NC), very efficiently internalized by tumor cells overexpressing the α isoform of the folate receptor; (ii) AS1411-linked nanocages (Apt-NC), internalized through nucleolin, a protein overexpressed in the cell surface of many types of cancers; and (iii) nanostructures that harbor both folate and AS1411 aptamer functionalization (Fol-Apt-NC). We analyzed the specific miRNA silencing activity of all types of nanostructures harboring miRNA sequestering sequences complementary to miR-21 and the cytotoxic effect when loaded with doxorubicin in a drug-resistant triple-negative breast cancer cell line. We demonstrate that the presence of folate as a targeting ligand increases the efficiency in miR-21 silencing compared to nanocages functionalized with AS1411. Double-functionalized nanocages (Fol-Apt-NC), loaded with doxorubicin, resulted in an increase of over 51% of the cytotoxic effect on MDA-MB-231 cells compared to free doxorubicin, demonstrating, besides selectivity, the ability of nanocages to overcome Dox chemoresistance. The higher efficiency of the folate-functionalized nanocages is due to the way of entrance, which induces more than four times higher intracellular stability and indicates that the folate-mediated route of cell entry is more efficient than the nucleolin-mediated one when both folate and AS1411 modifications are present.
Collapse
Affiliation(s)
- Valeria Unida
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giulia Vindigni
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Sofia Raniolo
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Alessandro Desideri
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Silvia Biocca
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-06-72-596-418
| |
Collapse
|
10
|
Allen NC, Chauhan R, Bates PJ, O’Toole MG. Optimization of Tumor Targeting Gold Nanoparticles for Glioblastoma Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3869. [PMID: 36364644 PMCID: PMC9653665 DOI: 10.3390/nano12213869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Glioblastoma brain tumors represent an aggressive form of gliomas that is hallmarked by being extremely invasive and aggressive due to intra and inter-tumoral heterogeneity. This complex tumor microenvironment makes even the newer advancements in glioblastoma treatment less effective long term. In developing newer treatment technologies against glioblastoma, one should tailor the treatment to the tumor microenvironment, thus allowing for a more robust and sustained anti-glioblastoma effect. Here, we present a novel gold nanoparticle therapy explicitly designed for bioactivity against glioblastoma representing U87MG cell lines. We employ standard conjugation techniques to create oligonucleotide-coated gold nanoparticles exhibiting strong anti-glioblastoma behavior and optimize their design to maximize bioactivity against glioblastoma. Resulting nanotherapies are therapy specific and show upwards of 75% inhibition in metabolic and proliferative activity with stark effects on cellular morphology. Ultimately, these gold nanotherapies are a good base for designing more multi-targeted approaches to fighting against glioblastoma.
Collapse
Affiliation(s)
- Nicholas C. Allen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Rajat Chauhan
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Paula J. Bates
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Martin G. O’Toole
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
11
|
Kashikar R, Kotha AK, Shah S, Famta P, Singh SB, Srivastava S, Chougule MB. Advances in nanoparticle mediated targeting of RNA binding protein for cancer. Adv Drug Deliv Rev 2022; 185:114257. [PMID: 35381306 DOI: 10.1016/j.addr.2022.114257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/28/2022] [Accepted: 03/30/2022] [Indexed: 12/24/2022]
Abstract
RNA binding proteins (RBPs) enact a very crucial part in the RNA directive processes. Atypical expression of these RBPs affects many steps of RNA metabolism, majorly altering its expression. Altered expression and dysfunction of RNA binding proteins lead to cancer progression and other diseases. We enumerate various available interventions, and recent findings focused on targeting RBPs for cancer therapy and diagnosis. The treatment, sensitization, chemoprevention, gene-mediated, and virus mediated interventions were studied to treat and diagnose cancer. The application of passively and actively targeted lipidic nanoparticles, polymeric nanoparticles, virus-based particles, and vaccine-based immunotherapy for the delivery of therapeutic agent/s against cancer are discussed. We also discuss the formulation aspect of nanoparticles for achieving delivery at the site of action and ongoing clinical trials targeting RBPs.
Collapse
|
12
|
Yang Y, Zheng X, Chen L, Gong X, Yang H, Duan X, Zhu Y. Multifunctional Gold Nanoparticles in Cancer Diagnosis and Treatment. Int J Nanomedicine 2022; 17:2041-2067. [PMID: 35571258 PMCID: PMC9094645 DOI: 10.2147/ijn.s355142] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/20/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is the second leading cause of death in the world, behind only cardiovascular diseases, and is one of the most serious diseases threatening human health nowadays. Cancer patients’ lives are being extended by the use of contemporary medical technologies, such as surgery, radiotherapy, and chemotherapy. However, these treatments are not always effective in extending cancer patients’ lives. Simultaneously, these approaches are often accompanied with a series of negative consequences, such as the occurrence of adverse effects and an increased risk of relapse. As a result, the development of a novel cancer-eradication strategy is still required. The emergence of nanomedicine as a promising technology brings a new avenue for the circumvention of limitations of conventional cancer therapies. Gold nanoparticles (AuNPs), in particular, have garnered extensive attention due to their many specific advantages, including customizable size and shape, multiple and useful physicochemical properties, and ease of functionalization. Based on these characteristics, many therapeutic and diagnostic applications of AuNPs have been exploited, particularly for malignant tumors, such as drug and nucleic acid delivery, photodynamic therapy, photothermal therapy, and X-ray-based computed tomography imaging. To leverage the potential of AuNPs, these applications demand a comprehensive and in-depth overview. As a result, we discussed current achievements in AuNPs in anticancer applications in a more methodical manner in this review. Also addressed in depth are the present status of clinical trials, as well as the difficulties that may be encountered when translating some basic findings into the clinic, in order to serve as a reference for future studies.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Xi Zheng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Lu Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Xuefeng Gong
- POWERCHINA Chengdu Engineering Corporation Limited, Chengdu, 611130, People’s Republic of China
| | - Hao Yang
- POWERCHINA Chengdu Engineering Corporation Limited, Chengdu, 611130, People’s Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Yuxuan Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
- Correspondence: Yuxuan Zhu, Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China, Email
| |
Collapse
|
13
|
Choo P, Arenas-Esteban D, Jung I, Chang WJ, Weiss EA, Bals S, Odom TW. Investigating Reaction Intermediates during the Seedless Growth of Gold Nanostars Using Electron Tomography. ACS NANO 2022; 16:4408-4414. [PMID: 35239309 DOI: 10.1021/acsnano.1c10669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Good's buffers can act both as nucleating and shape-directing agents during the synthesis of anisotropic gold nanostars (AuNS). Although different Good's buffers can produce AuNS shapes with branches that are oriented along specific crystallographic directions, the mechanism is not fully understood. This paper reports how an analysis of the intermediate structures during AuNS synthesis from HEPES, EPPS, and MOPS Good's buffers can provide insight into the formation of seedless AuNS. Electron tomography of AuNS structures quenched at early times (minutes) was used to characterize the morphology of the incipient seeds, and later times were used to construct the growth maps. Through this approach, we identified how the crystallinity and shape of the first structures synthesized with different Good's buffers determine the final AuNS morphologies.
Collapse
Affiliation(s)
- Priscilla Choo
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Daniel Arenas-Esteban
- EMAT, University of Antwerp, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
- NANOlab Center of Excellence, University of Antwerp, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Insub Jung
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Woo Je Chang
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Emily A Weiss
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Sara Bals
- EMAT, University of Antwerp, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
- NANOlab Center of Excellence, University of Antwerp, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Teri W Odom
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
14
|
Zare I, Yaraki MT, Speranza G, Najafabadi AH, Haghighi AS, Nik AB, Manshian BB, Saraiva C, Soenen SJ, Kogan MJ, Lee JW, Apollo NV, Bernardino L, Araya E, Mayer D, Mao G, Hamblin MR. Gold nanostructures: synthesis, properties, and neurological applications. Chem Soc Rev 2022; 51:2601-2680. [PMID: 35234776 DOI: 10.1039/d1cs01111a] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances in technology are expected to increase our current understanding of neuroscience. Nanotechnology and nanomaterials can alter and control neural functionality in both in vitro and in vivo experimental setups. The intersection between neuroscience and nanoscience may generate long-term neural interfaces adapted at the molecular level. Owing to their intrinsic physicochemical characteristics, gold nanostructures (GNSs) have received much attention in neuroscience, especially for combined diagnostic and therapeutic (theragnostic) purposes. GNSs have been successfully employed to stimulate and monitor neurophysiological signals. Hence, GNSs could provide a promising solution for the regeneration and recovery of neural tissue, novel neuroprotective strategies, and integrated implantable materials. This review covers the broad range of neurological applications of GNS-based materials to improve clinical diagnosis and therapy. Sub-topics include neurotoxicity, targeted delivery of therapeutics to the central nervous system (CNS), neurochemical sensing, neuromodulation, neuroimaging, neurotherapy, tissue engineering, and neural regeneration. It focuses on core concepts of GNSs in neurology, to circumvent the limitations and significant obstacles of innovative approaches in neurobiology and neurochemistry, including theragnostics. We will discuss recent advances in the use of GNSs to overcome current bottlenecks and tackle technical and conceptual challenges.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | | | - Giorgio Speranza
- CMM - FBK, v. Sommarive 18, 38123 Trento, Italy.,IFN - CNR, CSMFO Lab., via alla Cascata 56/C Povo, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, v. Sommarive 9, 38123 Trento, Italy
| | - Alireza Hassani Najafabadi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alireza Shourangiz Haghighi
- Department of Mechanical Engineering, Shiraz University of Technology, Modarres Boulevard, 13876-71557, Shiraz, Iran
| | - Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Cláudia Saraiva
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg.,Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, 8380492 Santiago, Chile
| | - Jee Woong Lee
- Department of Medical Sciences, Clinical Neurophysiology, Uppsala University, Uppsala, SE-751 23, Sweden
| | - Nicholas V Apollo
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,School of Physics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marques d'Avila e Bolama, 6201-001 Covilha, Portugal
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Av. Republica 275, Santiago, Chile
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Germany
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Michael R Hamblin
- Laser Research Center, University of Johannesburg, Doorfontein 2028, South Africa.
| |
Collapse
|
15
|
Tong X, Ga L, Ai J, Wang Y. Progress in cancer drug delivery based on AS1411 oriented nanomaterials. J Nanobiotechnology 2022; 20:57. [PMID: 35101048 PMCID: PMC8805415 DOI: 10.1186/s12951-022-01240-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/02/2022] [Indexed: 02/07/2023] Open
Abstract
Targeted cancer therapy has become one of the most important medical methods because of the spreading and metastatic nature of cancer. Based on the introduction of AS1411 and its four-chain structure, this paper reviews the research progress in cancer detection and drug delivery systems by modifying AS1411 aptamers based on graphene, mesoporous silica, silver and gold. The application of AS1411 in cancer treatment and drug delivery and the use of AS1411 as a targeting agent for the detection of cancer markers such as nucleoli were summarized from three aspects of active targeting, passive targeting and targeted nucleic acid apharmers. Although AS1411 has been withdrawn from clinical trials, the research surrounding its structural optimization is still very popular. Further progress has been made in the modification of nanoparticles loaded with TCM extracts by AS1411.
Collapse
Affiliation(s)
- Xin Tong
- College of Chemistry and Environmental Science, College of Geographical Science, Inner Mongolia Key Laboratory of Environmental Chemistry, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot, 010022, China
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot, 010110, China
| | - Jun Ai
- College of Chemistry and Environmental Science, College of Geographical Science, Inner Mongolia Key Laboratory of Environmental Chemistry, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot, 010022, China.
| | - Yong Wang
- College of Chemistry and Environmental Science, College of Geographical Science, Inner Mongolia Key Laboratory of Environmental Chemistry, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot, 010022, China.
| |
Collapse
|
16
|
Behroozi Z, Rahimi B, Kookli K, Safari MS, Hamblin MR, Razmgir M, Janzadeh A, Ramezani F. Distribution of gold nanoparticles into the brain: a systematic review and meta-analysis. Nanotoxicology 2021; 15:1059-1072. [PMID: 34591733 DOI: 10.1080/17435390.2021.1966116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite the widespread use of gold nanoparticles (GNPs), there is no consensus on their distribution to different tissues and organs. The present systematic review and meta-analysis addresses the accumulation of GNPs in brain tissue. Extensive searches were conducted in electronic databases, Medline, Web of Science, EMBASE, and Scopus. Based on inclusion and exclusion criteria, primary and secondary screening was performed. The value of brain accumulation of gold nanoparticle (the percentage of the injection dose of GNPs/gram of brain tissue that applied as effect size (ES) in analysis) and the standard error of the mean were extracted from articles and analyzed by calculating the pooled ES and the pooled confidence interval (CI) using STATA software. p ≤ 0.05 was considered significant. Thirty-eight studies were included in the meta-analysis. The results showed that the amount of GNPs was 0.06% of the injection dose/gram of brain tissue (ES = 0.06, %95 CI: 0.06-0.06, p < 0.0001). Considering the time between injection and tissue harvest (follow-up time), after 1 h the GNPs in brain tissue was 0.288% of the injection dose/gram of tissue (ES = 0.29, 95% CI: 0.25-0.33, p < 0.0001), while after four weeks it was only 0.02% (ES = 0.02, 95% CI: 0.01-0.03, p < 0.0001) of the injection dose/gram of tissue. The amount of GNPs in brain tissue was higher for PEG-coated GNPs compared to uncoated GNPs, and it was 5.6 times higher for rod-shaped GNPs compared to spherical GNPs. The mean amount of GNPs in the brain tissues of animals bearing a tumor was 5.8 times higher than in normal animals.
Collapse
Affiliation(s)
- Zahra Behroozi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Rahimi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Keihan Kookli
- International campus, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad S Safari
- Veterinary Faculty of Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Maryam Razmgir
- Medical Librarianship and Information Science, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Siegel AL, Baker GA. Bespoke nanostars: synthetic strategies, tactics, and uses of tailored branched gold nanoparticles. NANOSCALE ADVANCES 2021; 3:3980-4004. [PMID: 36132836 PMCID: PMC9417963 DOI: 10.1039/d0na01057j] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/20/2021] [Indexed: 05/05/2023]
Abstract
Interest in branched colloidal gold nanosystems has gained increased traction due to the structures' outstanding optical and plasmonic properties, resulting in utilization in techniques such as surface-enhanced spectroscopy and bioimaging, as well as plasmon photocatalysis and photothermal therapy. The unique morphologies of nanostars, multipods, urchins, and other highly branched nanomaterials exhibit selective optical and crystallographic features accessible by alterations in the respective wet-chemical syntheses, opening a vast array of useful applications. Examination of discriminatory reaction conditions, such as seeded growth (e.g., single-crystalline vs. multiply twinned seeds), underpotential deposition of Ag(i), galvanic replacement, and the dual use of competing reducing and capping agents, is shown to reveal conditions necessary for the genesis of assorted branched nanoscale gold frameworks. By observing diverse approaches, including template-directed, microwave-mediated, and aggregation-based methods, among others, a schema of synthetic pathways can be constructed to provide a guiding roadmap for obtaining the full range of desired branched gold nanocrystals. This review presents a comprehensive summary of such advances and these nuances of the underlying procedures, as well as offering mechanistic insights into the directed nanoscale growth. We conclude the review by discussing various applications for these fascinating nanomaterials, particularly surface-enhanced Raman spectroscopy, photothermal and photodynamic therapy, catalysis, drug delivery, and biosensing.
Collapse
Affiliation(s)
- Asher L Siegel
- Department of Chemistry, University of Missouri-Columbia Columbia MO 65211 USA
| | - Gary A Baker
- Department of Chemistry, University of Missouri-Columbia Columbia MO 65211 USA
| |
Collapse
|
18
|
Lopes-Nunes J, Oliveira PA, Cruz C. G-Quadruplex-Based Drug Delivery Systems for Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:671. [PMID: 34358097 PMCID: PMC8308530 DOI: 10.3390/ph14070671] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
G-quadruplexes (G4s) are a class of nucleic acids (DNA and RNA) with single-stranded G-rich sequences. Owing to the selectivity of some G4s, they are emerging as targeting agents to overtake side effects of several potential anticancer drugs, and delivery systems of small molecules to malignant cells, through their high affinity or complementarity to specific targets. Moreover, different systems are being used to improve their potential, such as gold nano-particles or liposomes. Thus, the present review provides relevant data about the different studies with G4s as drug delivery systems and the challenges that must be overcome in the future research.
Collapse
Affiliation(s)
- Jéssica Lopes-Nunes
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal;
| | - Paula A. Oliveira
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Carla Cruz
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal;
| |
Collapse
|
19
|
Liao J, Tian T, Shi S, Xie X, Peng S, Zhu Y, Xiao J, Lin Y. Broadening the biocompatibility of gold nanorods from rat to Macaca fascicularis: advancing clinical potential. J Nanobiotechnology 2021; 19:195. [PMID: 34193184 PMCID: PMC8243831 DOI: 10.1186/s12951-021-00941-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/21/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The biomedical field has used gold nanorods (GNRs) for decades; however, clinical trials and translation is limited except gold nanoshells. The preparation of gold nanoshells is more complex than that of polyethylene glycol-modified GNRs (PEG-GNRs), and it is difficult to ensure uniform thickness. It is important to encourage and broaden the use of the star member (PEG-GNRs) of gold nanoparticles family for clinical translation. Existing studies on PEG-GNRs are limited with no relevant systematic progression in non-human primates. Herein, we assessed the systematic biocompatibility of PEG-GNRs in rats and clinically relevant Macaca fascicularis. RESULTS In this small animal study, we administrated multiple doses of PEG-GNRs to rats and observed good biocompatibility. In the non-human primate study, PEG-GNRs had a longer blood half-life and produced a negligible immune response. Histological analysis revealed no significant abnormality. CONCLUSIONS PEG-GNRs were well-tolerated with good biocompatibility in both small animals and large non-human primates. The information gained from the comprehensive systemic toxicity assessment of PEG-GNRs in M. fascicularis will be helpful for translation to clinical trials.
Collapse
Affiliation(s)
- Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Taorang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xueping Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuanglin Peng
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Synchrotron Radiation Facility, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.
- Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jingang Xiao
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
20
|
Kozani PS, Kozani PS, Malik MT. AS1411-functionalized delivery nanosystems for targeted cancer therapy. EXPLORATION OF MEDICINE 2021; 2:146-166. [PMID: 34723284 PMCID: PMC8555908 DOI: 10.37349/emed.2021.00039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/27/2021] [Indexed: 12/12/2022] Open
Abstract
Nucleolin (NCL) is a multifunctional nucleolar phosphoprotein harboring critical roles in cells such as cell proliferation, survival, and growth. The dysregulation and overexpression of NCL are related to various pathologic and oncological indications. These characteristics of NCL make it an ideal target for the treatment of various cancers. AS1411 is a synthetic quadruplex-forming nuclease-resistant DNA oligonucleotide aptamer which shows a considerably high affinity for NCL, therefore, being capable of inducing growth inhibition in a variety of tumor cells. The high affinity and specificity of AS1411 towards NCL make it a suitable targeting tool, which can be used for the functionalization of therapeutic payloaddelivery nanosystems to selectively target tumor cells. This review explores the advances in NCL-targeting cancer therapy through AS1411-functionalized delivery nanosystems for the selective delivery of a broad spectrum of therapeutic agents.
Collapse
Affiliation(s)
- Pooria Safarzadeh Kozani
- Carlos Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115/111, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht 41446/66949, Iran
- Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht 41446/66949, Iran
| | - Mohammad Tariq Malik
- Departments of Microbiology and Immunology, Regenerative Medicine, and Stem Cell Biology, University of Louisville, Louisville, KY 40202, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
21
|
Jindal M, Nagpal M, Singh M, Aggarwal G, Dhingra GA. Gold Nanoparticles- Boon in Cancer Theranostics. Curr Pharm Des 2021; 26:5134-5151. [PMID: 32611300 DOI: 10.2174/1381612826666200701151403] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/23/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cancer is the world's second-largest cause of death, with an estimated 9.6 million fatalities in 2018. Malignant tumour (cancer) is caused by a mixture of genetic modifications due to the environmental variables that tend to activate or inactivate different genes, ultimately resulting in neoplastic transformations. Cancer is a multi-stage process that results from the conversion of the ordinary cells to tumour cells and progresses from a pre-cancer lesion to abnormal growth. METHODS Chemotherapy inhibits the ability of the cells to divide rapidly in an abnormal manner, but this treatment simultaneously affects the entire cellular network in the human body leading to cytotoxic effects. In this review article, the same issue has been addressed by discussing various aspects of the newer class of drugs in cancer therapeutics, i.e., Gold Nanoparticles (AuNPs) from metal nanoparticle (NP) class. RESULTS Metal NPs are advantageous over conventional chemotherapy as the adverse drug reactions are lesser. Additionally, ease of drug delivery, targeting and gene silencing are salient features of this treatment. Functionalized ligand-targeting metal NPs provide better energy deposition control in tumour. AuNPs are promising agents in the field of cancer treatment and are comprehensively studied as contrast agents, carriers of medicinal products, radiosensitizers and photothermal agents. For the targeted delivery of chemotherapeutic agents, AuNPs are used and also tend to enhance tumour imaging in vivo for a variety of cancer types and diseased organs. CONCLUSION The first part of the review focuses on various nano-carriers that are used for cancer therapy and deals with the progression of metal NPs in cancer therapy. The second part emphasizes the use of nanotechnology by considering the latest studies for diagnostic and therapeutic properties of AuNPs. AuNPs present the latest studies in the field of nanotechnology, which leads to the development of early-stage clinical trials. The next part of the review discusses the major features of five principal types of AuNPs: gold nanorods, gold nanoshells, gold nanospheres, gold nanocages, and gold nanostars that have their application in photothermal therapy (PTT).
Collapse
Affiliation(s)
- Mehak Jindal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Geeta Aggarwal
- Delhi Pharmaceutical Sciences and Research University, New Delhi-110017, India
| | | |
Collapse
|
22
|
Choo P, Liu T, Odom TW. Nanoparticle Shape Determines Dynamics of Targeting Nanoconstructs on Cell Membranes. J Am Chem Soc 2021; 143:4550-4555. [PMID: 33735562 DOI: 10.1021/jacs.1c00850] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanoparticle carriers are effective drug delivery vehicles. Along with other design parameters including size, composition, and surface charge, particle shape strongly influences cellular uptake. How nanoparticle geometry affects targeted delivery under physiologically relevant conditions, however, is inconclusive. Here, we demonstrate that nanoconstruct core shape influences the dynamics of targeting ligand-receptor interactions on cancer cell membranes. By single-particle tracking of translational and rotational motion, we compared DNA aptamer AS1411 conjugated gold nanostars (AS1411-AuNS) and 50 nm gold spheres (AS1411-50NPs) on cells with and without targeted nucleolin membrane receptors. On nucleolin-expressing cells, AS1411-AuNS exhibited faster velocities under directed diffusion and translated over larger areas during restricted diffusion compared to AS1411-50NPs, despite their similar protein corona profiles. On nucleolin-inhibited cells, AS1411-AuNS showed faster rotation dynamics over smaller translational areas, while AS1411-50NPs did not display significant changes in translation. These differences in translational and rotational motions indicate that nanoparticle shape affects how targeting nanoconstructs bind to cell-membrane receptors.
Collapse
|
23
|
Ni S, Zhuo Z, Pan Y, Yu Y, Li F, Liu J, Wang L, Wu X, Li D, Wan Y, Zhang L, Yang Z, Zhang BT, Lu A, Zhang G. Recent Progress in Aptamer Discoveries and Modifications for Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9500-9519. [PMID: 32603135 DOI: 10.1021/acsami.0c05750] [Citation(s) in RCA: 324] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Aptamers are oligonucleotide sequences with a length of about 25-80 bases which have abilities to bind to specific target molecules that rival those of monoclonal antibodies. They are attracting great attention in diverse clinical translations on account of their various advantages, including prolonged storage life, little batch-to-batch differences, very low immunogenicity, and feasibility of chemical modifications for enhancing stability, prolonging the half-life in serum, and targeted delivery. In this Review, we demonstrate the emerging aptamer discovery technologies in developing advanced techniques for producing aptamers with high performance consistently and efficiently as well as requiring less cost and resources but offering a great chance of success. Further, the diverse modifications of aptamers for therapeutic applications including therapeutic agents, aptamer-drug conjugates, and targeted delivery materials are comprehensively summarized.
Collapse
Affiliation(s)
- Shuaijian Ni
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Zhenjian Zhuo
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yufei Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuanyuan Yu
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Fangfei Li
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Jin Liu
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Luyao Wang
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Xiaoqiu Wu
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Dijie Li
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Youyang Wan
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Aiping Lu
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| | - Ge Zhang
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong 999077, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong 999077, China
- HKBU and IncreasePharm Joint Centre for Nucleic Acid Drug Discovery, Hong Kong 999077, China
| |
Collapse
|
24
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
25
|
Morita K, Nishimura Y, Nakamura S, Arai Y, Numako C, Sato K, Nakayama M, Akasaka H, Sasaki R, Ogino C, Kondo A. Titanium oxide nano-radiosensitizers for hydrogen peroxide delivery into cancer cells. Colloids Surf B Biointerfaces 2020; 198:111451. [PMID: 33223346 DOI: 10.1016/j.colsurfb.2020.111451] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/16/2020] [Accepted: 11/03/2020] [Indexed: 11/18/2022]
Abstract
Polyacrylic acid-modified titanium peroxide nanoparticles (PAA-TiOx NPs) are promising radiosensitizers that enhance the therapeutic effect of X-ray irradiation after local injection into tumors. However, the mechanism for this reaction has remained unclear with the exception of the involvement of hydrogen peroxide (H2O2), which is released by PAA-TiOx NPs to a liquid phase during dispersion. In the present study, a clonogenic assay was used to compare PAA-TiOx NPs with free H2O2 molecules to investigate the effect exerted on the radiosensitivity of cancer cells in vitro. A cell-free dialysis method revealed that a portion of the H2O2 adsorbed onto the PAA-TiOx NPs during synthesis could be released during a treatment regimen. The H2O2 release lasted for 7 h, which was sufficient for one radiation treatment procedure. For in vitro experiments, cultured human pancreatic cancer cells took up PAA-TiOx NPs in 10 min after administration. Interestingly, when the cells were washed with a buffer after treatment with either a PAA-TiOx NP or H2O2 solution, the intracellular H2O2 levels remained higher with PAA-TiOx NP treatment compared with the H2O2 solution treatment. Furthermore, the effects of subsequent X-ray irradiation corresponded to the intracellular H2O2 levels. These results indicate that PAA-TiOx NPs are efficient carriers of H2O2 into cancer cells and thus enhance the radiosensitivity.
Collapse
Affiliation(s)
- Kenta Morita
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan; Research Facility Center for Science and Technology, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Yuya Nishimura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Satoko Nakamura
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| | - Yuki Arai
- Graduate School of Science, Chiba University, 1-33, Yayoi, Inage, Chiba, 263-8522, Japan
| | - Chiya Numako
- Graduate School of Science, Chiba University, 1-33, Yayoi, Inage, Chiba, 263-8522, Japan
| | - Kazuyoshi Sato
- Division of Environmental Engineering Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjincho, Kiryu, Gunma, 376-8515, Japan
| | - Masao Nakayama
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kusunokicho 7-5-2, Chuou-ku, Kobe 650-0017, Japan
| | - Chiaki Ogino
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan.
| | - Akihiko Kondo
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan; Organization of Advanced Science and Technology, Kobe University, Rokkodaicho 1-1, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
26
|
Gonçalves ASC, Rodrigues CF, Moreira AF, Correia IJ. Strategies to improve the photothermal capacity of gold-based nanomedicines. Acta Biomater 2020; 116:105-137. [PMID: 32911109 DOI: 10.1016/j.actbio.2020.09.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/21/2022]
Abstract
The plasmonic photothermal properties of gold nanoparticles have been widely explored in the biomedical field to mediate a photothermal effect in response to the irradiation with an external light source. Particularly, in cancer therapy, the physicochemical properties of gold-based nanomaterials allow them to efficiently accumulate in the tumor tissue and then mediate the light-triggered thermal destruction of cancer cells with high spatial-temporal control. Nevertheless, the gold nanomaterials can be produced with different shapes, sizes, and organizations such as nanospheres, nanorods, nanocages, nanoshells, and nanoclusters. These gold nanostructures will present different plasmonic photothermal properties that can impact cancer thermal ablation. This review analyses the application of gold-based nanomaterials in cancer photothermal therapy, emphasizing the main parameters that affect its light-to-heat conversion efficiency and consequently the photothermal potential. The different shapes/organizations (clusters, shells, rods, stars, cages) of gold nanomaterials and the parameters that can be fine-tuned to improve the photothermal capacity are presented. Moreover, the gold nanostructures combination with other materials (e.g. silica, graphene, and iron oxide) or small molecules (e.g. indocyanine green and IR780) to improve the nanomaterials photothermal capacity is also overviewed.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Carolina F Rodrigues
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - André F Moreira
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Ilídio J Correia
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CIEPQF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal.
| |
Collapse
|
27
|
Valle AC, Yeh C, Huang Y. Near Infrared-Activatable Platinum-Decorated Gold Nanostars for Synergistic Photothermal/Ferroptotic Therapy in Combating Cancer Drug Resistance. Adv Healthc Mater 2020; 9:e2000864. [PMID: 32945131 DOI: 10.1002/adhm.202000864] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/15/2020] [Indexed: 12/15/2022]
Abstract
Ferroptotic cell death results from glutathione peroxidase 4 (GPX4) inactivation and/or glutathione (GSH) depletion. Elevated GSH levels are often found in multidrug-resistant (MDR) tumor cells, reducing their sensitivity to chemotherapeutic drugs and the efficacy of treatment. MDR cells also acquire a dependency on GPX4, reducing their oxidative stress and promoting their survival. Therefore, the depletion of GSH and inactivation of GPX4 has the potential to be a superior treatment strategy for MDR tumors. Platinum-decorated gold nanostars (Pt-AuNS) are presented as a novel metal nanoprodrug for ferroptotic therapy against MDR tumors. Under dark conditions, the synthesized Pt-AuNS exhibit negligible levels of toxicity. Upon exposure of the Pt-AuNS to near-infrared (NIR) light, active metallic (Pt and Au) species are released, subsequently inducing cytotoxicity. The mechanism of action is attributed to GSH depletion and GPX4 inactivation, accumulating lipid hydroperoxides, which in turn leads to ferroptosis. In in vivo xenograft, the MDR cancer model confirmed the NIR light-activation of Pt-AuNS prodrugs, resulting in efficient ferroptotic therapeutic action against MDR tumors without long-term side effects. The findings lay the groundwork for using Pt-AuNS prodrugs responsive to NIR light as ferroptosis-inducing agents in chemo-resistant cancer cells and demonstrate their potential for use in future clinical applications.
Collapse
Affiliation(s)
- Andrea C. Valle
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Chih‐Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| | - Yu‐Fen Huang
- Department of Biomedical Engineering and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
- Institute of Analytical and Environmental Sciences National Tsing Hua University Hsinchu Taiwan 30013 R.O.C
| |
Collapse
|
28
|
Light stimulus responsive nanomedicine in the treatment of oral squamous cell carcinoma. Eur J Med Chem 2020; 199:112394. [DOI: 10.1016/j.ejmech.2020.112394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/13/2022]
|
29
|
Combinatorial Inhibition of Cell Surface Receptors Using Dual Aptamer-Functionalized Nanoconstructs for Cancer Treatment. Pharmaceutics 2020; 12:pharmaceutics12070689. [PMID: 32708267 PMCID: PMC7408393 DOI: 10.3390/pharmaceutics12070689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Membrane receptors overexpressed in diseased states are considered novel therapeutic targets. However, the single targeting approach faces several fundamental issues, such as poor efficacy, resistance, and toxicity. Here, we report a dual-targeting strategy to enhance anti-cancer efficacy via synergistic proximity interactions between therapeutics and two receptor proteins. Importantly, we report the first finding of an interaction between c-Met and nucleolin and demonstrate the therapeutic value of targeting the interaction between them. Bispecific nanocarriers densely grafted with anti-c-Met and -nucleolin aptamer increased the local concentration of aptamers at the target sites, in addition to inducing target receptor clustering. It was also demonstrated that the simultaneous targeting of c-Met and nucleolin inhibited the cellular functions of the receptors and increased anti-cancer efficacy by altering the cell cycle. Our findings pave the way for the development of an effective combinatorial treatment based on nanoconstruct-mediated interaction between receptors.
Collapse
|
30
|
Yazdian-Robati R, Bayat P, Oroojalian F, Zargari M, Ramezani M, Taghdisi SM, Abnous K. Therapeutic applications of AS1411 aptamer, an update review. Int J Biol Macromol 2020; 155:1420-1431. [PMID: 31734366 DOI: 10.1016/j.ijbiomac.2019.11.118] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Nucleolin or C23, is one of the most abundant non-ribosomal phosphoproteins of nucleolus. However, in several cancers, nucleolin is highly expressed both intracellularly and on the cell surface. So, it is considered as a potential target for the diagnosis and cancer therapy. Targeting nucleolin by compounds such as AS1411 aptamer can reduce tumor cell growth. In this regard, interest has increased in nucleolin as a molecular target for overcoming cancer therapy challenges. This review paper addressed recent progresses in nucleolin targeting by the G-rich AS1411 aptamer in the field of cancer therapy mainly over the past three years.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Payam Bayat
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehryar Zargari
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Del Valle AC, Su CK, Sun YC, Huang YF. NIR-cleavable drug adducts of gold nanostars for overcoming multidrug-resistant tumors. Biomater Sci 2020; 8:1934-1950. [PMID: 32039412 DOI: 10.1039/c9bm01813a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
An aptamer-conjugated gold nanostar (dsDDA-AuNS) has been developed for targeting nucleolin present in both tumor cells and tumor vasculature for conducting a drug-resistant cancer therapy. AuNS with its strong absorption in the near-infrared (NIR) region was assembled with a layer of the anti-nucleolin aptamer AS1411. An anticancer drug, namely doxorubicin (DOX), was specifically conjugated on deoxyguanosine residues employing heat and acid labile methylene linkages. In response to NIR irradiation, dsDDA-AuNS allowed on-demand therapeutics. AS1411 played an active role in drug cargo-nucleus interactions, enhancing drug accumulation in the nuclei of drug-resistant breast cancer cells. The intravenous injection of dsDDA-AuNS allowed higher drug accumulation in drug-resistant tumors over naked drugs, leading to greater therapeutic efficacy even at a 54-fold less equivalent drug dose. The in vivo triggered release of DOX from dsDDA-AuNS was achieved by NIR irradiation, resulting in simultaneous photothermal and chemotherapeutic actions, yielding superior tumor growth inhibition than those obtained from either type of monotherapy for overcoming drug resistance in cancers.
Collapse
Affiliation(s)
- Andrea C Del Valle
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China.
| | - Cheng-Kuan Su
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, 20224, Taiwan, Republic of China
| | - Yuh-Chang Sun
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China. and Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China
| | - Yu-Fen Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China. and Institute of Analytical and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013 Taiwan, Republic of China
| |
Collapse
|
32
|
Cheng YH, He C, Riviere JE, Monteiro-Riviere NA, Lin Z. Meta-Analysis of Nanoparticle Delivery to Tumors Using a Physiologically Based Pharmacokinetic Modeling and Simulation Approach. ACS NANO 2020; 14:3075-3095. [PMID: 32078303 PMCID: PMC7098057 DOI: 10.1021/acsnano.9b08142] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/20/2020] [Indexed: 05/18/2023]
Abstract
Numerous studies have engineered nanoparticles with different physicochemical properties to enhance the delivery efficiency to solid tumors, yet the mean and median delivery efficiencies are only 1.48% and 0.70% of the injected dose (%ID), respectively, according to a study using a nonphysiologically based modeling approach based on published data from 2005 to 2015. In this study, we used physiologically based pharmacokinetic (PBPK) models to analyze 376 data sets covering a wide range of nanomedicines published from 2005 to 2018 and found mean and median delivery efficiencies at the last sampling time point of 2.23% and 0.76%ID, respectively. Also, the mean and median delivery efficiencies were 2.24% and 0.76%ID at 24 h and were decreased to 1.23% and 0.35%ID at 168 h, respectively, after intravenous administration. While these delivery efficiencies appear to be higher than previous findings, they are still quite low and represent a critical barrier in the clinical translation of nanomedicines. We explored the potential causes of this poor delivery efficiency using the more mechanistic PBPK perspective applied to a subset of gold nanoparticles and found that low delivery efficiency was associated with low distribution and permeability coefficients at the tumor site (P < 0.01). We also demonstrate how PBPK modeling and simulation can be used as an effective tool to investigate tumor delivery efficiency of nanomedicines.
Collapse
Affiliation(s)
- Yi-Hsien Cheng
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Jim E. Riviere
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- 1Data
Consortium, Kansas State University, Manhattan, Kansas 66506, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhoumeng Lin
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
33
|
Mousavi SM, Zarei M, Hashemi SA, Ramakrishna S, Chiang WH, Lai CW, Gholami A. Gold nanostars-diagnosis, bioimaging and biomedical applications. Drug Metab Rev 2020; 52:299-318. [PMID: 32150480 DOI: 10.1080/03602532.2020.1734021] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gold Nanostars (GNS) have attracted tremendous attention toward themselves owing to their multi-branched structure and unique properties. These state of the art metallic nanoparticles possess intrinsic features like remarkable optical properties and exceptional physiochemical activities. These star-shaped gold nanoparticles can predominantly be utilized in biosensing, photothermal therapy, imaging, surface-enhanced Raman spectroscopy and target drug delivery applications due to their low toxicity and extraordinary optical features. In the current review, recent approaches in the matter of GNS in case of diagnosis, bioimaging and biomedical applications were summarized and reported. In this regard, first an overview about the structure and general properties of GNS were reported and thence detailed information regarding the diagnostic, bioimaging, photothermal therapy, and drug delivery applications of such novel nanomaterials were presented in detail. Summarized information clearly highlighting the superior capability of GNS as potential multi-functional materials for biomedical applications.
Collapse
Affiliation(s)
- Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Maryam Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Alireza Hashemi
- Department of Mechanical Engineering, Center for Nanofibers and Nanotechnology, National University of Singapore, Singapore, Singapore
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, Center for Nanofibers and Nanotechnology, National University of Singapore, Singapore, Singapore
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Chin Wei Lai
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Ahmad Gholami
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Pharmaceutical research Center, Shiraz University of Medical Science, Shiraz, Iran
| |
Collapse
|
34
|
Hu B, Berkey C, Feliciano T, Chen X, Li Z, Chen C, Amini S, Nai MH, Lei QL, Ni R, Wang J, Leow WR, Pan S, Li YQ, Cai P, Miserez A, Li S, Lim CT, Wu YL, Odom TW, Dauskardt RH, Chen X. Thermal-Disrupting Interface Mitigates Intercellular Cohesion Loss for Accurate Topical Antibacterial Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1907030. [PMID: 32072703 PMCID: PMC7702719 DOI: 10.1002/adma.201907030] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/12/2020] [Indexed: 05/10/2023]
Abstract
Bacterial infections remain a leading threat to global health because of the misuse of antibiotics and the rise in drug-resistant pathogens. Although several strategies such as photothermal therapy and magneto-thermal therapy can suppress bacterial infections, excessive heat often damages host cells and lengthens the healing time. Here, a localized thermal managing strategy, thermal-disrupting interface induced mitigation (TRIM), is reported, to minimize intercellular cohesion loss for accurate antibacterial therapy. The TRIM dressing film is composed of alternative microscale arrangement of heat-responsive hydrogel regions and mechanical support regions, which enables the surface microtopography to have a significant effect on disrupting bacterial colonization upon infrared irradiation. The regulation of the interfacial contact to the attached skin confines the produced heat and minimizes the risk of skin damage during thermoablation. Quantitative mechanobiology studies demonstrate the TRIM dressing film with a critical dimension for surface features plays a critical role in maintaining intercellular cohesion of the epidermis during photothermal therapy. Finally, endowing wound dressing with the TRIM effect via in vivo studies in S. aureus infected mice demonstrates a promising strategy for mitigating the side effects of photothermal therapy against a wide spectrum of bacterial infections, promoting future biointerface design for antibacterial therapy.
Collapse
Affiliation(s)
- Benhui Hu
- Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Christopher Berkey
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Timothy Feliciano
- Department of Materials Science and Engineering and Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Xiaohong Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Zhuyun Li
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Chao Chen
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Shahrouz Amini
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Mui Hoon Nai
- Department of Biomedical Engineering, Mechanobiology Institute, Institute for Health Innovation and Technology (iHealthtech) National University of Singapore, 9 Engineering Drive 1, Singapore, 117576, Singapore
| | - Qun-Li Lei
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Ran Ni
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore, 637459, Singapore
| | - Juan Wang
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Wan Ru Leow
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Shaowu Pan
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Yong-Qiang Li
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Pingqiang Cai
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Ali Miserez
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Shuzhou Li
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, Mechanobiology Institute, Institute for Health Innovation and Technology (iHealthtech) National University of Singapore, 9 Engineering Drive 1, Singapore, 117576, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Teri W Odom
- Department of Materials Science and Engineering and Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
| | - Reinhold H Dauskardt
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Xiaodong Chen
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
35
|
Hu B, Wang J, Li J, Li S, Li H. Superiority of L-tartaric Acid Modified Chiral Mesoporous Silica Nanoparticle as a Drug Carrier: Structure, Wettability, Degradation, Bio-Adhesion and Biocompatibility. Int J Nanomedicine 2020; 15:601-618. [PMID: 32099354 PMCID: PMC6996211 DOI: 10.2147/ijn.s233740] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/15/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE The purpose of this research was to study the basic physicochemical and biological properties regarding the application of L-tartaric acid modified chiral mesoporous silica nanoparticle (CMSN) as a drug carrier, and to explore the structure-property relationship of silica-based materials. METHODS CMSN with functions of carboxyl modification and chirality was successfully synthesized through co-condensation method, and the basic characteristics of CMSN, including morphology, structure, wettability, degradation, bio-adhesion and retention ability in gastrointestinal tract (GI tract) were estimated by comparing with non-functionalized mesoporous silica nanoparticles (MSN). Meanwhile, the biocompatibility and toxicity of L-tartaric modification were systematically evaluated both in vitro and in vivo through MTT cell viability assay, cell cycle and apoptosis assay, hemolysis assay, histopathology examination, hematology analysis, and clinical chemistry examination. RESULTS CMSN and MSN were spherical nanoparticles with uniform mesoporous structure. CMSN with smaller pore size and carboxyl functional groups exhibited better wettability. Besides, CMSN and MSN could dissolve thoroughly in simulated physiological fluids during a degradation period of 1-12 weeks. Interestingly, the in vitro and in vivo behaviors of carriers, including degradation, bio-adhesion and retention ability in the GI tract were closely related to wettability. As expected, CMSN had faster degradation rate, higher mucosa-adhesion ability, and longer retention time. Particularly, CMSN improved the bio-adhesion property in both gastric mucosa and small intestinal mucosa, and prolonged the GI tract retention time to at least 12 h, which meant higher probability for absorption. The biocompatibility and toxicity examination indicated that CMSN was a kind of biocompatible bio-material with good blood compatibility and negligible toxicity, which is required for further applications in biological fields. CONCLUSION CMSN with functions of carboxyl modification and chirality had superiority in terms of both physicochemical and biological properties. The in vitro and in vivo behaviors of carriers, including degradation, bio-adhesion, and retention were closely related to wettability.
Collapse
Affiliation(s)
- Beibei Hu
- School of Pharmacy, China Medical University, Shenyang110122, People’s Republic of China
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang050018, People’s Republic of China
| | - Jianxin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Jing Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Sanming Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang110016, People’s Republic of China
| | - Heran Li
- School of Pharmacy, China Medical University, Shenyang110122, People’s Republic of China
| |
Collapse
|
36
|
Wang J, Liang D, Jin Q, Feng J, Tang X. Bioorthogonal SERS Nanotags as a Precision Theranostic Platform for in Vivo SERS Imaging and Cancer Photothermal Therapy. Bioconjug Chem 2020; 31:182-193. [PMID: 31940174 DOI: 10.1021/acs.bioconjchem.0c00022] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Precise detection and effective treatment are crucial to prolong cancer patients' lives. Surface-enhanced Raman scattering (SERS) imaging coupled with photothermal therapy has been considered a precise and effective strategy for cancer theranostics. Nevertheless, Raman reporters employed in the literature usually possessed multiple shift peaks in the fingerprint region, which are overlapped with background signals from endogenous biological molecules. Herein, we fabricated a new kind of bioorthogonal Raman reporter and aptamer functionalized SERS nanotags. The SERS nanotags demonstrated a strong Raman signal at 2205 cm-1 in the biologically Raman-silent region and recognized MCF-7 breast cancer cells for Raman imaging with high specificity. Laser irradiation induced serious toxicity of MCF-7 cells due to the excellent photothermal capability of the SERS nanotags. After intravenous administration of the SERS nanotags, tumor Raman spectral detection and mapping in living mice were successfully achieved. Further in vivo antitumor experiments manifested that the aptamer-modified SERS nanotags significantly restrained tumor growth after laser irradiation with 99% inhibition rate and good biocompatibility. These results clearly revealed that the SERS nanotags could serve as a novel and precise theranostic platform for in vivo cancer diagnosis and photothermal therapy.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Peking University , No. 38, Xueyuan Rd. , Beijing 100191 , P.R. China
| | - Duanwei Liang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Peking University , No. 38, Xueyuan Rd. , Beijing 100191 , P.R. China
| | - Qingqing Jin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Peking University , No. 38, Xueyuan Rd. , Beijing 100191 , P.R. China
| | - Jie Feng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Peking University , No. 38, Xueyuan Rd. , Beijing 100191 , P.R. China
| | - Xinjing Tang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences , Peking University , No. 38, Xueyuan Rd. , Beijing 100191 , P.R. China
| |
Collapse
|
37
|
Kabirian-Dehkordi S, Chalabi-Dchar M, Mertani HC, Le Guellec D, Verrier B, Diaz JJ, Mehrgardi MA, Bouvet P. AS1411-conjugated gold nanoparticles affect cell proliferation through a mechanism that seems independent of nucleolin. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 21:102060. [PMID: 31336175 DOI: 10.1016/j.nano.2019.102060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/11/2019] [Accepted: 06/26/2019] [Indexed: 12/15/2022]
Abstract
G-rich oligonucleotide, AS1411, has been shown to interact with nucleolin and to inhibit cancer cell proliferation and tumor growth. This antiproliferative action is increased when AS1411 is conjugated to different types of nanoparticles. However, the molecular mechanisms are not known. In this work, we show in several cell lines that optimized AS1411-conjugated gold nanoparticles (GNS-AS1411) inhibit nucleolin expression at the RNA and protein levels. We observed an alteration of the nucleolar structure with a decrease of ribosomal RNA accumulation comparable to what is observed upon nucleolin knock down. However, the expression of genes involved in cell cycle and the cell cycle blockage by GNS-AS1411 are not regulated in the same way as that in cells where nucleolin has been knocked down. These data suggest that the anti-proliferative activity of GNS-AS1411 is not the only consequence of nucleolin targeting and down-regulation.
Collapse
Affiliation(s)
- Samaneh Kabirian-Dehkordi
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre, Léon Bérard, Lyon, France; Department of chemistry, University of Isfahan, Isfahan, Iran
| | - Mounira Chalabi-Dchar
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre, Léon Bérard, Lyon, France
| | - Hichem C Mertani
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre, Léon Bérard, Lyon, France
| | - Dominique Le Guellec
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Université Claude Bernard Lyon 1, Centre, National de la Recherche Scientifique (CNRS), Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, Université Claude Bernard Lyon 1, Centre, National de la Recherche Scientifique (CNRS), Lyon, France
| | - Jean-Jacques Diaz
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre, Léon Bérard, Lyon, France
| | | | - Philippe Bouvet
- Université de Lyon, Centre de Recherche en Cancérologie de Lyon, Cancer Cell Plasticity Department, UMR INSERM 1052 CNRS 5286, Centre, Léon Bérard, Lyon, France; Université de Lyon, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
38
|
Jauffred L, Samadi A, Klingberg H, Bendix PM, Oddershede LB. Plasmonic Heating of Nanostructures. Chem Rev 2019; 119:8087-8130. [PMID: 31125213 DOI: 10.1021/acs.chemrev.8b00738] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The absorption of light by plasmonic nanostructures and their associated temperature increase are exquisitely sensitive to the shape and composition of the structure and to the wavelength of light. Therefore, much effort is put into synthesizing novel nanostructures for optimized interaction with the incident light. The successful synthesis and characterization of high quality and biocompatible plasmonic colloidal nanoparticles has fostered numerous and expanding applications, especially in biomedical contexts, where such particles are highly promising for general drug delivery and for tomorrow's cancer treatment. We review the thermoplasmonic properties of the most commonly used plasmonic nanoparticles, including solid or composite metallic nanoparticles of various dimensions and geometries. Common methods for synthesizing plasmonic particles are presented with the overall goal of providing the reader with a guide for designing or choosing nanostructures with optimal thermoplasmonic properties for a given application. Finally, the biocompatibility and biological tolerance of structures are critically discussed along with novel applications of plasmonic nanoparticles in the life sciences.
Collapse
Affiliation(s)
| | - Akbar Samadi
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| | - Henrik Klingberg
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| | | | - Lene B Oddershede
- Niels Bohr Institute , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
39
|
Liu Y, Huang W, Xiong C, Huang Y, Chen BJ, Racioppi L, Chao N, Vo-Dinh T. Biodistribution and sensitive tracking of immune cells with plasmonic gold nanostars. Int J Nanomedicine 2019; 14:3403-3411. [PMID: 31190799 PMCID: PMC6514259 DOI: 10.2147/ijn.s192189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022] Open
Abstract
Aim: To quantitatively and sensitively investigate the biodistribution of immune cells after systemic administration. Methods: Immune cells were loaded with plasmonic gold nanostars (GNS) tracking probes. Inductively coupled plasma mass spectrometry (ICP-MS) was used for quantitative gold mass measurement and two-photon photoluminescence (TPL) was used for high-resolution sensitive optical imaging. Results: GNS nanoparticles were loaded successfully into immune cells without negative effect on cellular vitality. Liver and spleen were identified to be the major organs for macrophage cells uptake after systematic administration. A small amount of macrophage cells were detected in the tumor site in our murine lymphoma animal model. Conclusion: GNS has great potential as a biocompatible marker for quantitative tracking and high-resolution imaging of immune cells at the cellular level.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Wei Huang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Chuanfeng Xiong
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Yuxian Huang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Benny J Chen
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Luigi Racioppi
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | - Nelson Chao
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA.,Fitzpatrick Institute for Photonics, Duke University, Durham, NC 27708, USA.,Department of Chemistry, Duke University, Durham, NC 27708, USA
| |
Collapse
|
40
|
Li B, Lane LA. Probing the biological obstacles of nanomedicine with gold nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1542. [PMID: 30084539 PMCID: PMC6585966 DOI: 10.1002/wnan.1542] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Despite massive growth in nanomedicine research to date, the field still lacks fundamental understanding of how certain physical and chemical features of a nanoparticle affect its ability to overcome biological obstacles in vivo and reach its intended target. To gain fundamental understanding of how physical and chemical parameters affect the biological outcomes of administered nanoparticles, model systems that can systematically manipulate a single parameter with minimal influence on others are needed. Gold nanoparticles are particularly good model systems in this case as one can synthetically control the physical dimensions and surface chemistry of the particles independently and with great precision. Additionally, the chemical and physical properties of gold allow particles to be detected and quantified in tissues and cells with high sensitivity. Through systematic biological studies using gold nanoparticles, insights toward rationally designed nanomedicine for in vivo imaging and therapy can be obtained. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Bin Li
- Department of Biomedical Engineering, College of Engineering and Applied SciencesNanjing UniversityNanjingJiangsuChina
| | - Lucas A. Lane
- Department of Biomedical Engineering, College of Engineering and Applied SciencesNanjing UniversityNanjingJiangsuChina
| |
Collapse
|
41
|
Valcourt DM, Dang MN, Day ES. IR820-loaded PLGA nanoparticles for photothermal therapy of triple-negative breast cancer. J Biomed Mater Res A 2019; 107:1702-1712. [PMID: 30920169 DOI: 10.1002/jbm.a.36685] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/25/2019] [Accepted: 03/22/2019] [Indexed: 01/06/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for 15-25% of breast cancer cases and lacks expression of the three most common receptors seen on other subtypes of breast cancer. This lack of expression makes TNBC unsusceptible to currently available targeted or hormonal therapies, so new treatment strategies are desperately needed. Photothermal therapy (PTT), which utilizes nanoparticles (NPs) embedded in tumors as exogenous energy absorbers to convert externally applied near-infrared (NIR) light into heat to ablate cancer cells, has shown promise as an alternative strategy. However, it typically uses gold-based NPs that will remain in the body for extended period of time with unknown long-term health effects. To enable PTT with biodegradable, polymeric NPs, we encapsulated the NIR-absorbing dye IR820 in poly(lactic-co-glycolic acid) (PLGA) NPs. We characterized the physicochemical properties of these IR820-loaded PLGA NPs and evaluated their performance as PTT agents using both in vitro and in vivo models of TNBC. The results demonstrate that these NPs are potent mediators of PTT that induce cell death primarily through apoptosis to effectively hinder the growth of TNBC tumors. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1702-1712, 2019.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716.,Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware, 19716.,Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, Delaware, 19713
| |
Collapse
|
42
|
Wu R, Min Q, Guo J, Zheng T, Jiang L, Zhu JJ. Sequential Delivery and Cascade Targeting of Peptide Therapeutics for Triplexed Synergistic Therapy with Real-Time Monitoring Shuttled by Magnetic Gold Nanostars. Anal Chem 2019; 91:4608-4617. [DOI: 10.1021/acs.analchem.8b05877] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Rong Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jingjing Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Tingting Zheng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai 200241, China
| | - Liping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
43
|
Ma Y, Li W, Zhou Z, Qin X, Wang D, Gao Y, Yu Z, Yin F, Li Z. Peptide-Aptamer Coassembly Nanocarrier for Cancer Therapy. Bioconjug Chem 2019; 30:536-540. [PMID: 30702869 DOI: 10.1021/acs.bioconjchem.8b00903] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We reported methionine bis-alkylated nonapeptide Wpc as an efficient siRNA vehicle previously. Herein, we report an aptamer could also spontaneously coassemble with Wpc to form uniformed nanoparticles for efficient delivery. This unique peptide-based aptamer nanocarrier showed significantly improved cell penetration and antiproliferation effect with high biocompatibility toward various cancer cell lines.
Collapse
Affiliation(s)
- Yue Ma
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Wenjun Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Ziyuan Zhou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China.,Chemical Biology Laboratory for Infectious Diseases, State Key Discipline of Infectious Diseases , Shenzhen Third People's Hospital , Shenzhen 518020 , China
| | - Xuan Qin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Dongyuan Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Yubo Gao
- School of Information Engineering , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening , Southern Medical University , Guangzhou 510515 , China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| |
Collapse
|
44
|
Yan R, Chen J, Wang J, Rao J, Du X, Liu Y, Zhang L, Qiu L, Liu B, Zhao YD, Jiang P, Chen C, Li YQ. A NanoFlare-Based Strategy for In Situ Tumor Margin Demarcation and Neoadjuvant Gene/Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802745. [PMID: 30294858 DOI: 10.1002/smll.201802745] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/29/2018] [Indexed: 06/08/2023]
Abstract
Accurate tumor margin demarcation in situ remains a paramount challenge. Herein, a NanoFlare (also known as spherical-nucleic-acid technology) based strategy is reported for in situ tumor margin delineation by transforming and amplifying the pathophysiological redox signals of tumor microenvironment. The NanoFlare designed (named AuNS-ASON) is based on gold nanostar (AuNS) coated with a dense shell of disulfide bridge-inserted and cyanine dyes-labeled antisense oligonucleotides (ASON) targeting survivin mRNA. The unique anisotropic ASON-spike nanostructure endows the AuNS-ASON with universal cellular internalization of tumor cells, while the disulfide bridge inserted confers response specificity toward redox activation. In vitro experiments demonstrate that the AuNS-ASON can discriminate tumor cells rapidly with activated fluorescence signals (>100-fold) in 2 h, and further achieve synergistic gene/photothermal tumor cells ablation upon near-infrared laser irradiation. Remarkably, in situ tumor margin delineation with high accuracy and outstanding spatial resolution (<100 µm) in mice bearing different tumors is obtained based on the AuNS-ASON, providing intraoperative guidance for tumor resection. Moreover, the AuNS-ASON can enable efficient neoadjuvant gene/photothermal therapy before surgery to reduce tumor extent and increase resectability. The concept of NanoFlare-based microenvironment signal transformation and amplification could be used as a general strategy to guide the design of activatable nanoprobes for cancer theranostics.
Collapse
Affiliation(s)
- Rong Yan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Jie Chen
- Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215002, China
| | - Jianhao Wang
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, 213164, China
| | - Jiaming Rao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Xuancheng Du
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, 213164, China
| | - Yongming Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Leshuai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Lin Qiu
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, 213164, China
| | - Bo Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yuan-Di Zhao
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Pengju Jiang
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, 213164, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Yong-Qiang Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| |
Collapse
|
45
|
Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents. CRYSTALS 2018. [DOI: 10.3390/cryst8100369] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Breast cancer (BC) is the most common cancer in women worldwide, with a mortality rate that has been forecasted to rise in the next decade. This is especially worrying for people with triple-negative BC (TNBC), because of its unresponsiveness to current therapies. Different drugs to treat TNBC have been assessed, and, although platinum chemotherapy drugs seem to offer some hope, their drawbacks have motivated extensive investigations into alternative metal-based BC therapies. This paper aims to: (i) describe the preliminary in vitro and in vivo anticancer properties of non-platinum metal-based complexes (NPMBC) against TNBC; and (ii) analyze the likely molecular targets involved in their anticancer activity.
Collapse
|
46
|
Yang PC, Wu T, Lin YW. Label-Free Colorimetric Detection of Mercury (II) Ions Based on Gold Nanocatalysis. SENSORS (BASEL, SWITZERLAND) 2018; 18:E2807. [PMID: 30149653 PMCID: PMC6163656 DOI: 10.3390/s18092807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 12/01/2022]
Abstract
Herein, a label-free colorimetric nanosensor for Hg(II) is developed utilizing the hindering effect of Hg(II) on the kinetic aspect of gold nanoparticle (AuNPs) growth on the surface of gold nanostars (AuNSs). H-AuNS probes are synthesized and modified by 2-[4-(2-hydroxyethel) piperazine-1-yl] ethanesulfonic acid (HEPES). After the formulation of the reagents and testing conditions are optimized, HEPES-capped AuNSs (H-AuNSs) demonstrates good selectivity and sensitivity towards Hg(II) determination. A H-AuNS probe, in the presence of HCl/Au(III)/H₂O₂, is capable of detecting a Hg(II) concentration range of 1.0 nM⁻100 µM, with a detection limit of 0.7 nM, at a signal-to-noise ratio of 3.0, and a visual detection limit of 10 nM with naked eyes. For practicality, the H-AuNS probe is evaluated by measuring Hg(II) in the environmental water matrices (lake water and seawater) by a standard addition and recovery study. The detection limits for environmental samples are found to be higher than the lab samples, but they are still within the maximum allowable Hg concentration in drinking water (10 nM) set by the US Environmental Protection Agency (EPA). To create a unique nanosensor, the competitive interaction between Hg(II) and Pt(IV) toward the H-AuNSs probe is developed into a logic gate, improving the specificity in the detection of Hg(II) ions in water samples.
Collapse
Affiliation(s)
- Pei-Chia Yang
- Department of Chemistry, National Changhua University of Education, 1, Jin-De Road, Changhua City 50007, Taiwan.
| | - Tsunghsueh Wu
- Department of Chemistry, University of Wisconsin-Platteville, 1 University Plaza, Platteville, WI 53818-3099, USA.
| | - Yang-Wei Lin
- Department of Chemistry, National Changhua University of Education, 1, Jin-De Road, Changhua City 50007, Taiwan.
| |
Collapse
|
47
|
Larue L, Ben Mihoub A, Youssef Z, Colombeau L, Acherar S, André JC, Arnoux P, Baros F, Vermandel M, Frochot C. Using X-rays in photodynamic therapy: an overview. Photochem Photobiol Sci 2018; 17:1612-1650. [DOI: 10.1039/c8pp00112j] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Photodynamic therapy is a therapeutic option to treat cancer and other diseases.
Collapse
|
48
|
Zhuo Z, Yu Y, Wang M, Li J, Zhang Z, Liu J, Wu X, Lu A, Zhang G, Zhang B. Recent Advances in SELEX Technology and Aptamer Applications in Biomedicine. Int J Mol Sci 2017; 18:2142. [PMID: 29036890 PMCID: PMC5666824 DOI: 10.3390/ijms18102142] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
Aptamers are short DNA/RNA oligonucleotides capable of binding to target molecules with high affinity and specificity. The process of selecting an aptamer is called Systematic Evolution of Ligands by Exponential Enrichment (SELEX). Thanks to the inherit merits, aptamers have been used in a wide range of applications, including disease diagnosis, targeted delivery agents and therapeutic uses. To date, great achievements regarding the selection, modifications and application of aptamers have been made. However, few aptamer-based products have already successfully entered into clinical and industrial use. Besides, it is still a challenge to obtain aptamers with high affinity in a more efficient way. Thus, it is important to comprehensively review the current shortage and achievement of aptamer-related technology. In this review, we first present the limitations and notable advances of aptamer selection. Then, we compare the different methods used in the kinetic characterization of aptamers. We also discuss the impetus and developments of the clinical application of aptamers.
Collapse
Affiliation(s)
- Zhenjian Zhuo
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Maolin Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Jie Li
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Zongkang Zhang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Xiaohao Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Institute of Precision Medicine and Innovative Drug Discovery, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Shenzhen Lab of Combinatorial Compounds and Targeted Drug Delivery, HKBU Institute of Research and Continuing Education, Shenzhen 518000, China.
| | - Baoting Zhang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
49
|
Liu Y, Maccarini P, Palmer GM, Etienne W, Zhao Y, Lee CT, Ma X, Inman BA, Vo-Dinh T. Synergistic Immuno Photothermal Nanotherapy (SYMPHONY) for the Treatment of Unresectable and Metastatic Cancers. Sci Rep 2017; 7:8606. [PMID: 28819209 PMCID: PMC5561243 DOI: 10.1038/s41598-017-09116-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/07/2017] [Indexed: 01/05/2023] Open
Abstract
Metastatic spread is the mechanism in more than 90 percent of cancer deaths and current therapeutic options, such as systemic chemotherapy, are often ineffective. Here we provide a proof of principle for a novel two-pronged modality referred to as Synergistic Immuno Photothermal Nanotherapy (SYMPHONY) having the potential to safely eradicate both primary tumors and distant metastatic foci. Using a combination of immune-checkpoint inhibition and plasmonic gold nanostar (GNS)–mediated photothermal therapy, we were able to achieve complete eradication of primary treated tumors and distant untreated tumors in some mice implanted with the MB49 bladder cancer cells. Delayed rechallenge with MB49 cancer cells injection in mice that appeared cured by SYMPHONY did not lead to new tumor formation after 60 days observation, indicating that SYMPHONY treatment induced effective long-lasting immunity against MB49 cancer cells.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.,Department of Chemistry, Duke University, Durham, NC, 27708, USA
| | - Paolo Maccarini
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Gregory M Palmer
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Wiguins Etienne
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yulin Zhao
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Chen-Ting Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Xiumei Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Brant A Inman
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA. .,Department of Chemistry, Duke University, Durham, NC, 27708, USA. .,Fitzpatrick Institute of Photonics, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
50
|
Riley RS, Day ES. Gold nanoparticle-mediated photothermal therapy: applications and opportunities for multimodal cancer treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1449. [PMID: 28160445 PMCID: PMC5474189 DOI: 10.1002/wnan.1449] [Citation(s) in RCA: 407] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 11/04/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022]
Abstract
Photothermal therapy (PTT), in which nanoparticles embedded within tumors generate heat in response to exogenously applied laser light, has been well documented as an independent strategy for highly selective cancer treatment. Gold-based nanoparticles are the main mediators of PTT because they offer: (1) biocompatibility, (2) small diameters that enable tumor penetration upon systemic delivery, (3) simple gold-thiol bioconjugation chemistry for the attachment of desired molecules, (4) efficient light-to-heat conversion, and (5) the ability to be tuned to absorb near-infrared light, which penetrates tissue more deeply than other wavelengths of light. In addition to acting as a standalone therapy, gold nanoparticle-mediated PTT has recently been evaluated in combination with other therapies, such as chemotherapy, gene regulation, and immunotherapy, for enhanced anti-tumor effects. When delivered independently, the therapeutic success of molecular agents is hindered by premature degradation, insufficient tumor delivery, and off-target toxicity. PTT can overcome these limitations by enhancing tumor- or cell-specific delivery of these agents or by sensitizing cancer cells to these additional therapies. All together, these benefits can enhance the therapeutic success of both PTT and the secondary treatment while lowering the required doses of the individual agents, leading to fewer off-target effects. Given the benefits of combining gold nanoparticle-mediated PTT with other treatment strategies, many exciting opportunities for multimodal cancer treatment are emerging that will ultimately lead to improved patient outcomes. WIREs Nanomed Nanobiotechnol 2017, 9:e1449. doi: 10.1002/wnan.1449 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Rachel S. Riley
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Emily S. Day
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
| |
Collapse
|