1
|
Fernandes DA. Multifunctional gold nanoparticles for cancer theranostics. 3 Biotech 2024; 14:267. [PMID: 39416669 PMCID: PMC11473483 DOI: 10.1007/s13205-024-04086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
The diagnosis and treatment of cancer can often be challenging requiring more attractive options. Some types of cancers are more aggressive than others and symptoms for many cancers are subtle, especially in the early stages. Nanotechnology provides high sensitivity, specificity and multimodal capability for cancer detection, treatment and monitoring. In particular, metal nanoparticles (NPs) such as gold nanoparticles (AuNPs) are attractive nanosystems for researchers interested in bioimaging and therapy. The size, shape and surface of AuNPs can be modified for improving targeting and accumulation in cancer cells, for example through introduction of ligands and surface charge. The interactions of AuNPs with electromagnetic radiation (e.g., visible-near-infrared, X-rays) can be used for photothermal therapy and radiation therapy, through heat generated from light absorption and emission of Auger electrons, respectively. The subsequent expansion and high X-ray attenuation from AuNPs can be used for enhancing contrast for tumor detection (e.g., using photoacoustic, computed tomography imaging). Multi-functionality can be further extended through covalent/non-covalent functionalization, for loading additional imaging/therapeutic molecules for combination therapy and multimodal imaging. In order to cover the important aspects for designing and using AuNPs for cancer theranostics, this review focuses on the synthesis, functionalization and characterization methods that are important for AuNPs, and presents their unique properties and different applications in cancer theranostics.
Collapse
|
2
|
Tao C, Yu N, Ren Q, Wen M, Qiu P, Niu S, Li M, Chen Z. Dressing and undressing MOF nanophotosensitizers to manipulate phototoxicity for precise therapy of tumors. Acta Biomater 2024; 177:444-455. [PMID: 38325709 DOI: 10.1016/j.actbio.2024.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
Photodynamic therapy (PDT) is a clinically approved treatment for tumors, and it relies on the phototoxicity of photosensitizers by producing reactive oxygen species (ROS) to destroy cancer cells under light irradiation. However, such phototoxicity is a double-edged sword, which is also harmful to normal tissues. To manipulate phototoxicity and improve the therapy effect, herein we have proposed a dressing-undressing strategy for de-activating and re-activating therapy functions of photosensitizer nanoparticles. One kind of metal organic framework (PCN-224), which is composed of Zr(IV) cation and tetrakis (4-carboxyphenyl) porphyrin (TCPP), has been prepared as a model of photosensitizer, and it has size of ∼70 nm. These PCN-224 nanoparticles are subsequently coated with a mesoporous organic silica (MOS) shell containing tetrasulfide bonds (-S-S-S-S-), realizing the dressing of PCN-224. MOS shell has the thickness of ∼20 nm and thus can block 1O2 (diffusion distance: <10 nm), deactivating the phototoxicity and preventing the damage to skin and eyes. Furthermore, PCN-224@MOS can be used to load chemotherapy drug (DOX·HCl). When PCN-224@MOS-DOX are mixed with glutathione (GSH), MOS shell with -S-S-S-S- bonds can be reduced by GSH and then be decomposed, which results in the undressing and then confers the exposure of PCN-224 with good PDT function as well as the release of DOX. When PCN-224@MOS-DOX dispersion is injected into the mice and accumulated in the tumor, endogenous GSH also confers the undressing of PCN-224@MOS-DOX, realizing the in-situ activation of PDT and chemotherapy for tumor. Therefore, the present study not only demonstrates a general dressing-undressing strategy for manipulating phototoxicity of photosensitizers, but also provide some insights for precise therapy of tumors without side-effects. STATEMENT OF SIGNIFICANCE: Photosensitizers can generate reactive oxygen species (ROS) under light radiation to destroy cancer cells. However, this phototoxicity is a double-edged sword and also harmful to normal tissues such as the skin and eyes. To control phototoxicity and improve therapeutic efficacy, we prepared a PCN-224@MOS-DOX nanoplatform and proposed a dressing and undressing strategy to deactivate and reactivate the therapeutic function of the photosensitizer nanoparticles. The MOS shell can block the diffusion of 1O2, eliminate phototoxicity, and prevent damage to the skin and eyes. When injected into mice and accumulated in tumors, PCN-224@MOS-DOX dispersions are endowed with an endogenous GSH-driven undressing effect, achieving in situ activation of PDT and tumor chemotherapy.
Collapse
Affiliation(s)
- Cheng Tao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Nuo Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Qian Ren
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Mei Wen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Pu Qiu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Shining Niu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200072, China.
| | - Zhigang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
3
|
Truong DH, Tran PTT, Tran TH. Nanoparticles as carriers of photosensitizers to improve photodynamic therapy in cancer. Pharm Dev Technol 2024; 29:221-235. [PMID: 38407140 DOI: 10.1080/10837450.2024.2322570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Photodynamic therapy (PDT) has emerged as a promising non invasive therapeutic approach for cancer treatment, offering unique advantages over conventional treatments. The combination of light activation and photosensitizing agents allows for targeted and localized destruction of cancer cells, reducing damage to surrounding healthy tissues. In recent years, the integration of nanoparticles with PDT has garnered significant attention due to their potential to enhance therapeutic outcomes. This review article aims to provide a comprehensive overview of the current state-of-the-art in utilizing nanoparticles for photodynamic therapy in cancer treatment. We summarized various nanoparticle-based approaches, their properties, and their implications in optimizing PDT efficacy, and discussed challenges and prospects in the field.
Collapse
Affiliation(s)
| | - Phuong Thi Thu Tran
- Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Hanoi, Vietnam
| |
Collapse
|
4
|
Lei L, Dai W, Man J, Hu H, Jin Q, Zhang B, Tang Z. Lonidamine liposomes to enhance photodynamic and photothermal therapy of hepatocellular carcinoma by inhibiting glycolysis. J Nanobiotechnology 2023; 21:482. [PMID: 38102658 PMCID: PMC10724989 DOI: 10.1186/s12951-023-02260-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), has great promise in the treatment of cancer. However, there are many obstacles that can restrict the therapeutic efficacy of phototherapy. The hypoxic tumor microenvironment can restrict the production of reactive oxygen species (ROS) in PDT. As for PTT, the thermotolerance of cancer cells may lead to ineffective PTT. In this study, IR780 and glycolysis inhibitor lonidamine (LND)-encapsulated liposomes are prepared for photodynamic and photothermal therapy of hepatocellular carcinoma. IR780 can be used as a photosensitizer and photothermal agent for simultaneous PDT and PTT after being irradiated with 808 nm laser. LND can reduce the oxygen consumption of cancer cells by inhibiting glycolysis, which will relieve tumor hypoxia and produce more ROS for PDT. On the other hand, energy supply can be blocked by LND-induced glycolysis inhibition, which will inhibit the production of heat shock proteins (HSPs), reduce the thermotolerance of tumor cells, and finally enhance the therapeutic efficacy of PTT. The enhanced PTT is studied by measuring intracellular HSPs, ATP level, and mitochondrial membrane potential. The antitumor effect of IR780 and LND co-loaded liposomes is extensively investigated by in vitro and in vivo experiments. This research provides an innovative strategy to simultaneously enhance the therapeutic efficacy of PDT and PTT by inhibiting glycolysis, which is promising for future creative approaches to cancer phototherapy.
Collapse
Affiliation(s)
- Lei Lei
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiaping Man
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haitao Hu
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Bo Zhang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Yang A, Luo D, Jia Y, Liu Y, Zhang Z, Li S, Liu R, Zhou J, Wang J. Targeted delivery of AZD5363 to T-cell acute lymphocytic leukemia by mSiO 2-Au nanovehicles. Colloids Surf B Biointerfaces 2023; 230:113505. [PMID: 37574619 DOI: 10.1016/j.colsurfb.2023.113505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023]
Abstract
T-cell acute lymphocytic leukemia (T-ALL) is the most common cancer in children, with a low survival rate because of drug resistance and a high recurrence rate. Targeted delivery of chemotherapy drugs can reduce their side effects and improve their efficacy. The abnormality of phosphatidylinositol-3-kinase/protein kinase B/ mammalian target of rapamycin (PI3K/Akt/mTOR) pathway plays a key role in T-ALL occurrence. AZD5363 is a selective Akt inhibitor with promising therapeutic potential for tumors encoded by the PI3K/Akt/mTOR pathway. However, the toxicity and side effects have limited its application in treating T-ALL. This study aimed to design a delivery system for targeting AZD5363 to T-ALL by sgc8c aptamer designed as mesoporous silica (mSiO2) decorated with Au nanoparticles. The cell-specific targeting and cytotoxicity of mSiO2-Au-AZD5363-Apt were investigated. The mSiO2-Au nanovehicles were found feasible for AZD5363 delivery, with high loading efficiency and pH-responsive release in the acidic lysosome. More importantly, mSiO2-Au-AZD5363-Apt nanovehicles could specifically recognize and enter T-ALL cells in vitro and in vivo, effectively inhibiting the proliferation of CCRF-CEM cells. In conclusion, mSiO2-Au-AZD5363-Apt provided an effective therapeutic method for the targeted treatment of T-ALL.
Collapse
Affiliation(s)
- Aiyun Yang
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Danqing Luo
- Department of Pediatric Hematology Oncology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Yuxuan Jia
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Yuxin Liu
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Zuo Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Shen Li
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Rong Liu
- Department of Pediatric Hematology Oncology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jing Zhou
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Jianhua Wang
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
6
|
Arellano LG, Villar-Alvarez EM, Velasco B, Domínguez-Arca V, Prieto G, Cambón A, Barbosa S, Taboada P. Light excitation of gold Nanorod-Based hybrid nanoplatforms for simultaneous bimodal phototherapy. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
7
|
Yang J, Jia L, He Z, Wang Y. Recent advances in SN-38 drug delivery system. Int J Pharm 2023; 637:122886. [PMID: 36966982 DOI: 10.1016/j.ijpharm.2023.122886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
DNA topoisomerase I plays a key role in lubricatingthe wheels of DNA replication or RNA transcription through breaking and reconnecting DNA single-strand. It is widely known that camptothecin and its derivatives (CPTs) have inhibitory effects on topoisomerases I, and have obtained some clinical benefits in cancer treatment. The potent cytotoxicity makes 7-ethyl-10-hydroxycamptothecin (SN-38) become a brilliant star among these derivatives. However, some undesirable physical and chemical properties of this compound, including poor solubility and stability, seriously hinder its effective delivery to tumor sites. In recent years, strategies to alleviate these defects have aroused extensive research interest. By focusing on the loading mechanism, basic nanodrug delivery systems with SN-38 loaded, like nanoparticles, liposomes and micelles, are demonstrated here. Additionally, functionalized nanodrug delivery systems of SN-38 including prodrug and active targeted nanodrug delivery systems and delivery systems designed to overcome drug resistance are also reviewed. At last, challenges for future research in formulation development and clinical translation of SN-38 drug delivery system are discussed.
Collapse
|
8
|
Yan HH, Huang M, Zhu F, Cheng R, Wen S, Li LT, Liu H, Zhao XH, Luo FK, Huang CZ, Wang J. Two-Dimensional Analysis Method for Highly Sensitive Detection of Dual MicroRNAs in Breast Cancer Cells. Anal Chem 2023; 95:3968-3975. [PMID: 36792543 DOI: 10.1021/acs.analchem.2c03479] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Multiple biomarker detection is crucial for early clinical diagnosis, and it is significant to achieve the simultaneous detection of multiple biomarkers with the same nanomaterial. In this work, the hairpin DNA strands were selectively modified on the surface of gold nanorods (AuNRs) to construct two kinds of nanoprobes by rational design. When in the presence of dual microRNAs, AuNRs were assembled to form end-to-end (ETE) and side-by-side (SBS) dimers. Compared with a single AuNR, the dark-field scattering intensity and red color percentage variation of dimers were extremely distinguished, which could be developed for dual microRNA detection by combining the red color percentage and scattering intensity with the data processing method of principal component analysis to construct a two-dimensional analysis method. Especially, the fraction of AuNR dimers presented a linear relationship with the amount of microRNAs. Based on this, microRNA-21 and microRNA Let-7a in breast cancer cells were detected with the detection limits of 1.72 and 0.53 fM, respectively. This method not only achieved the sensitive detection of dual microRNAs in human serum but also realized the high-resolution intracellular imaging, which developed a new way for the oriented assembly of nanomaterials and biological detection in living cells.
Collapse
Affiliation(s)
- Hui Hong Yan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Min Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Fu Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Ru Cheng
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | | | - Liang Tong Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Hui Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Xiao Hui Zhao
- The Ninth People's Hospital of Chongqing, No. 69 Jialing Village, Beibei District, Chongqing 400700, China
| | - Fu Kang Luo
- The Ninth People's Hospital of Chongqing, No. 69 Jialing Village, Beibei District, Chongqing 400700, China
| | - Cheng Zhi Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| | - Jian Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P. R. China
| |
Collapse
|
9
|
Khademi R, Mohammadi Z, Khademi R, Saghazadeh A, Rezaei N. Nanotechnology-based diagnostics and therapeutics in acute lymphoblastic leukemia: a systematic review of preclinical studies. NANOSCALE ADVANCES 2023; 5:571-595. [PMID: 36756502 PMCID: PMC9890594 DOI: 10.1039/d2na00483f] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/19/2022] [Indexed: 05/23/2023]
Abstract
Background: Leukemia is a malignant disease that threatens human health and life. Nano-delivery systems improve drug solubility, bioavailability, and blood circulation time, and release drugs selectively at desired sites using targeting or sensing strategies. As drug carriers, they could improve therapeutic outcomes while reducing systemic toxicity. They have also shown promise in improving leukemia detection and diagnosis. The study aimed to assess the potential of nanotechnology-based diagnostics and therapeutics in preclinical human acute lymphoblastic leukemia (h-ALL). Methods: We performed a systematic search through April 2022. Articles written in English reporting the toxicity, efficacy, and safety of nanotechnology-based drugs (in the aspect of treatment) and specificity, limit of detection (LOD), or sensitivity (in the aspect of the detection field) in preclinical h-ALL were included. The study was performed according to PRISMA instructions. The methodological quality was assessed using the QualSyst tool. Results: A total of 63 original articles evaluating nanotechnology-based therapeutics and 35 original studies evaluating nanotechnology-based diagnostics were included in this review. As therapeutics in ALL, nanomaterials offer controlled release, targeting or sensing ligands, targeted gene therapy, photodynamic therapy and photothermic therapy, and reversal of multidrug-resistant ALL. A narrative synthesis of studies revealed that nanoparticles improve the ratio of efficacy to the toxicity of anti-leukemic drugs. They have also been developed as a vehicle for biomolecules (such as antibodies) that can help detect and monitor leukemic biomarkers. Therefore, nanomaterials can help with early diagnostics and personalized treatment of ALL. Conclusion: This review discussed nanotechnology-based preclinical strategies to achieve ALL diagnosis and therapy advancement. This involves modern drug delivery apparatuses and detection devices for prompt and targeted disease diagnostics. Nonetheless, we are yet in the experimental phase and investigational stage in the field of nanomedicine, with many features remained to be discovered as well as numerous problems to be solved.
Collapse
Affiliation(s)
- Reyhane Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN) Tehran Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN) Tehran Iran
- Department of Medical Laboratory Sciences, School of Para-medicine, Ahvaz Jundishapour University of Medical Sciences Ahvaz Iran
| | - Zahra Mohammadi
- Radiological Technology Department of Actually Paramedical Sciences, Babol University of Medical Sciences Babol Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN) Babol Iran
| | - Rahele Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN) Tehran Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences Dr Qarib St, Keshavarz Blvd Tehran 14194 Iran +98-21-6692-9235 +98-21-6692-9234
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences Dr Qarib St, Keshavarz Blvd Tehran 14194 Iran +98-21-6692-9235 +98-21-6692-9234
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN) Tehran Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
10
|
Alavi N, Maghami P, Pakdel AF, Rezaei M, Avan A. Antibody-modified Gold Nanobiostructures: Advancing Targeted Photodynamic Therapy for Improved Cancer Treatment. Curr Pharm Des 2023; 29:3103-3122. [PMID: 37990429 DOI: 10.2174/0113816128265544231102065515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/03/2023] [Indexed: 11/23/2023]
Abstract
Photodynamic therapy (PDT) is an innovative, non-invasive method of treating cancer that uses light-activated photosensitizers to create reactive oxygen species (ROS). However, challenges associated with the limited penetration depth of light and the need for precise control over photosensitizer activation have hindered its clinical translation. Nanomedicine, particularly gold nanobiostructures, offers promising solutions to overcome these limitations. This paper reviews the advancements in PDT and nanomedicine, focusing on applying antibody-modified gold nanobiostructures as multifunctional platforms for enhanced PDT efficacy and improved cancer treatment outcomes. The size, shape, and composition of gold nanobiostructures can significantly influence their PDT efficacy, making synthetic procedures crucial. Functionalizing the surface of gold nanobiostructures with various molecules, such as antibodies or targeting agents, bonding agents, PDT agents, photothermal therapy (PTT) agents, chemo-agents, immunotherapy agents, and imaging agents, allows composition modification. Integrating gold nanobiostructures with PDT holds immense potential for targeted cancer therapy. Antibody-modified gold nanobiostructures, in particular, have gained significant attention due to their tunable plasmonic characteristics, biocompatibility, and surface functionalization capabilities. These multifunctional nanosystems possess unique properties that enhance the efficacy of PDT, including improved light absorption, targeted delivery, and enhanced ROS generation. Passive and active targeting of gold nanobiostructures can enhance their localization near cancer cells, leading to efficient eradication of tumor tissues upon light irradiation. Future research and clinical studies will continue to explore the potential of gold nanobiostructures in PDT for personalized and effective cancer therapy. The synthesis, functionalization, and characterization of gold nanobiostructures, their interaction with light, and their impact on photosensitizers' photophysical and photochemical properties, are important areas of investigation. Strategies to enhance targeting efficiency and the evaluation of gold nanobiostructures in vitro and in vivo studies will further advance their application in PDT. The integrating antibody-modified gold nanobiostructures in PDT represents a promising strategy for targeted cancer therapy. These multifunctional nanosystems possess unique properties that enhance PDT efficacy, including improved light absorption, targeted delivery, and enhanced ROS generation. Continued research and development in this field will contribute to the advancement of personalized and effective cancer treatment approaches.
Collapse
Affiliation(s)
- Negin Alavi
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Parvaneh Maghami
- Department of Biology, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Azar Fani Pakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezaei
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane 4059, Australia
| |
Collapse
|
11
|
Functionalized Silver and Gold Nanomaterials with Diagnostic and Therapeutic Applications. Pharmaceutics 2022; 14:pharmaceutics14102182. [PMID: 36297620 PMCID: PMC9609291 DOI: 10.3390/pharmaceutics14102182] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
The functionalization of nanomaterials with suitable capping ligands or bioactive agents is an interesting strategy in designing nanosystems with suitable applicability and biocompatibility; the physicochemical and biological properties of these nanomaterials can be highly improved for biomedical applications. In this context, numerous explorations have been conducted in the functionalization of silver (Ag) and gold (Au) nanomaterials using suitable functional groups or agents to design nanosystems with unique physicochemical properties such as excellent biosensing capabilities, biocompatibility, targeting features, and multifunctionality for biomedical purposes. Future studies should be undertaken for designing novel functionalization tactics to improve the properties of Au- and Ag-based nanosystems and reduce their toxicity. The possible release of cytotoxic radicals or ions, the internalization of nanomaterials, the alteration of cellular signaling pathways, the translocation of these nanomaterials across the cell membranes into mitochondria, DNA damages, and the damage of cell membranes are the main causes of their toxicity, which ought to be comprehensively explored. In this study, recent advancements in diagnostic and therapeutic applications of functionalized Au and Ag nanomaterials are deliberated, focusing on important challenges and future directions.
Collapse
|
12
|
Theranostic Potentials of Gold Nanomaterials in Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14133047. [PMID: 35804818 PMCID: PMC9264814 DOI: 10.3390/cancers14133047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/03/2022] [Accepted: 06/17/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hematological malignancies (HMs) cover 50% of all malignancies, and people of all ages can be affected by these deadly diseases. In many cases, conventional diagnostic tools fail to diagnose HMs at an early stage, due to heterogeneity and the long-term indolent phase of HMs. Therefore, many patients start their treatment at the late stage of HMs and have poor survival. Gold nanomaterials (GNMs) have shown promise as a cancer theranostic agent. GNMs are 1 nm to 100 nm materials having magnetic resonance and surface-plasmon-resonance properties. GNMs conjugated with antibodies, nucleic acids, peptides, photosensitizers, chemotherapeutic drugs, synthetic-drug candidates, bioactive compounds, and other theranostic biomolecules may enhance the efficacy and efficiency of both traditional and advanced theranostic approaches to combat HMs. Abstract Hematological malignancies (HMs) are a heterogeneous group of blood neoplasia generally characterized by abnormal blood-cell production. Detection of HMs-specific molecular biomarkers (e.g., surface antigens, nucleic acid, and proteomic biomarkers) is crucial in determining clinical states and monitoring disease progression. Early diagnosis of HMs, followed by an effective treatment, can remarkably extend overall survival of patients. However, traditional and advanced HMs’ diagnostic strategies still lack selectivity and sensitivity. More importantly, commercially available chemotherapeutic drugs are losing their efficacy due to adverse effects, and many patients develop resistance against these drugs. To overcome these limitations, the development of novel potent and reliable theranostic agents is urgently needed to diagnose and combat HMs at an early stage. Recently, gold nanomaterials (GNMs) have shown promise in the diagnosis and treatment of HMs. Magnetic resonance and the surface-plasmon-resonance properties of GNMs have made them a suitable candidate in the diagnosis of HMs via magnetic-resonance imaging and colorimetric or electrochemical sensing of cancer-specific biomarkers. Furthermore, GNMs-based photodynamic therapy, photothermal therapy, radiation therapy, and targeted drug delivery enhanced the selectivity and efficacy of anticancer drugs or drug candidates. Therefore, surface-tuned GNMs could be used as sensitive, reliable, and accurate early HMs, metastatic HMs, and MRD-detection tools, as well as selective, potent anticancer agents. However, GNMs may induce endothelial leakage to exacerbate cancer metastasis. Studies using clinical patient samples, patient-derived HMs models, or healthy-animal models could give a precise idea about their theranostic potential as well as biocompatibility. The present review will investigate the theranostic potential of vectorized GNMs in HMs and future challenges before clinical theranostic applications in HMs.
Collapse
|
13
|
The restructure of Au@Ag nanorods for cell imaging with dark-field microscope. Talanta 2022; 244:123403. [PMID: 35349839 DOI: 10.1016/j.talanta.2022.123403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 11/22/2022]
Abstract
The facile and noninjurious image of cells with high resolution and low toxicity is essential since imaging can offer rich and direct information and insights into metabolic activities, clinical diagnosis, drug delivery and cancer therapy. In this contribution, a smart imaging probe was employed as a contrast agent for dark-field cell imaging. Au core/Ag shell nanorods (Au@Ag NRs) that characterized by X-ray diffraction and X-ray photoelectron spectroscopy, formed Au@Ag@AgI NRs when exposed to iodine, which greatly enhanced the light scattering of nanorods. Herein, the silver shell acted as the response element for iodine as well as the protective agent for Au core. When conjugated with folate, the nanorods can be used to image human cervical cancer cells (HeLa cells) under a dark-field microscope. Nanorods were demonstrated with excellent tumor cellular uptake ability without obvious cytotoxicity, making them ideal candidates in biosensing and bioimaging applications.
Collapse
|
14
|
Ci T, Zhang W, Qiao Y, Li H, Zang J, Li H, Feng N, Gu Z. Delivery strategies in treatments of leukemia. Chem Soc Rev 2022; 51:2121-2144. [PMID: 35188506 DOI: 10.1039/d1cs00755f] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukemia is a hematological malignancy associated with the uncontrolled proliferation of mutant progenitors, suppressing the production of normal blood cells. Current treatments, including chemotherapy, radiotherapy, and immunotherapy, still lead to unsatisfactory results with a 5 year survival rate of only 30-50%. The poor prognosis is related to both disease relapse and treatment-associated toxicity. Delivery strategies can improve the in vivo pharmacokinetics of drugs, navigating the therapeutics to target cells or the tumor microenvironment and reversing drug resistance, which maximizes tumor elimination and alleviates systematic adverse effects. This review discusses available FDA-approved anti-leukemia drugs and therapies with a focus on the advances in the development of anti-leukemia drug delivery systems. Additionally, challenges in clinical translation of the delivery strategies and future research opportunities in leukemia treatment are also included.
Collapse
Affiliation(s)
- Tianyuan Ci
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wentao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingyu Qiao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jing Zang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
15
|
Yu N, Qiu P, Ren Q, Wen M, Geng P, Macharia DK, Zhu M, Chen Z. Transforming a Sword into a Knife: Persistent Phototoxicity Inhibition and Alternative Therapeutical Activation of Highly-Photosensitive Phytochlorin. ACS NANO 2021; 15:19793-19805. [PMID: 34851096 DOI: 10.1021/acsnano.1c07241] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The phototoxicity of photosensitizers (PSs) is a double-edged sword with one edge beneficial for destroying tumors while the other is detrimental to normal tissues, and the conventional "OFF-ON" strategy provides temporary inhibition so that phototoxicity would come sooner or later due to the inevitable retention and transformation of PSs in vivo. We herein put forward a strategy to convert "double-edged sword" PSs into "single-edged knife" ones with simultaneously persistent phototoxicity inhibition and alternative multiple therapeutical activation. The Chlorin e6 (Ce6) as the PS model directly assembles with Cu2+ ions into nanoscale frameworks (nFs) whose Cu2+-coordination includes both carboxyl groups and a porphyrin ring of Ce6 instead of Fe3+/Mn2+-coordination with only carboxyl groups. Compared to the high phototoxicity of Ce6, the nFs exhibit efficient energy transfer due to the dual-coordination of paramagnetic Cu2+ ions and the aggregation, achieving the persistent and high phototoxicity inhibition rate of >92%. Alternatively, the nFs not only activate a high photoacoustic contrast and near-infrared (NIR)-driven photothermal efficacy (3.5-fold that of free Ce6) due to the aggregation-enhanced nonradiative transition but also initiate tumor microenvironment modulation, structure disassembly, and chemodynamic effect by Cu2+ ions. Given these merits, the nFs achieve long-term biosecurity, no retina injury under sunlight, and a higher therapeutical output than the photodynamic effect of Ce6. This work presents a possibility of converting numerous highly phototoxic porphyrins into safe and efficient ones.
Collapse
Affiliation(s)
- Nuo Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Pu Qiu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Qian Ren
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Mei Wen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Peng Geng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Daniel K Macharia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Zhigang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
16
|
Zheng J, Cheng X, Zhang H, Bai X, Ai R, Shao L, Wang J. Gold Nanorods: The Most Versatile Plasmonic Nanoparticles. Chem Rev 2021; 121:13342-13453. [PMID: 34569789 DOI: 10.1021/acs.chemrev.1c00422] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gold nanorods (NRs), pseudo-one-dimensional rod-shaped nanoparticles (NPs), have become one of the burgeoning materials in the recent years due to their anisotropic shape and adjustable plasmonic properties. With the continuous improvement in synthetic methods, a variety of materials have been attached around Au NRs to achieve unexpected or improved plasmonic properties and explore state-of-the-art technologies. In this review, we comprehensively summarize the latest progress on Au NRs, the most versatile anisotropic plasmonic NPs. We present a representative overview of the advances in the synthetic strategies and outline an extensive catalogue of Au-NR-based heterostructures with tailored architectures and special functionalities. The bottom-up assembly of Au NRs into preprogrammed metastructures is then discussed, as well as the design principles. We also provide a systematic elucidation of the different plasmonic properties associated with the Au-NR-based structures, followed by a discussion of the promising applications of Au NRs in various fields. We finally discuss the future research directions and challenges of Au NRs.
Collapse
Affiliation(s)
- Jiapeng Zheng
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Xizhe Cheng
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Han Zhang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Xiaopeng Bai
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Ruoqi Ai
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| | - Lei Shao
- Beijing Computational Science Research Center, Beijing 100193, China
| | - Jianfang Wang
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR 999077, China
| |
Collapse
|
17
|
Yan J, Gao T, Lu Z, Yin J, Zhang Y, Pei R. Aptamer-Targeted Photodynamic Platforms for Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:27749-27773. [PMID: 34110790 DOI: 10.1021/acsami.1c06818] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Achieving controlled and accurate delivery of photosensitizers (PSs) into tumor sites is a major challenge in conventional photodynamic therapy (PDT). Aptamer is a short oligonucleotide sequence (DNA or RNA) with a folded three-dimensional structure, which can selectively bind to specific small molecules, proteins, or the whole cells. Aptamers could act as ligands and be modified onto PSs or nanocarriers, enabling specific recognition and binding to tumor cells or their membrane proteins. The resultant aptamer-modified PSs or PSs-containing nanocarriers generate amounts of reactive oxygen species with light irradiation and obtain superior photodynamic therapeutic efficiency in tumors. Herein, we overview the recent progress in the designs and applications of aptamer-targeted photodynamic platforms for tumor therapy. First, we focus on the progress on the rational selection of aptamers and summarize the applications of aptamers which have been applied for targeted tumor diagnosis and therapy. Then, aptamer-targeted photodynamic therapies including various aptamer-PSs, aptamer-nanocarriers containing PSs, and aptamer-nano-photosensitizers are highlighted. The aptamer-targeted synergistically therapeutic platforms including PDT, photothermal therapy, and chemotherapy, as well as the imaging-guided theranostics, are also discussed. Finally, we offer an insight into the development trends and future perspectives of aptamer-targeted photodynamic platforms for tumor therapy.
Collapse
Affiliation(s)
- Jincong Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Tian Gao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Zhongzhong Lu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| |
Collapse
|
18
|
Zhang C, Chen W, Zhang T, Jiang X, Hu Y. Hybrid nanoparticle composites applied to photodynamic therapy: strategies and applications. J Mater Chem B 2021; 8:4726-4737. [PMID: 32104868 DOI: 10.1039/d0tb00093k] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic therapy (PDT), as a robust strategy, has long been applied to cancer treatment owing to the meaningful breakthroughs and unique advantages, including ignorable invasiveness and spatio-temporal selectivity. Numerous PDT agents, especially hybrid nanoparticle composite (hybrid)-based sensitizers consisting of an organic polymer and inorganic nanoparticles (NPs), feature the synergetic pros of the components, which have unlocked the additional potentials of PDT. Although reviews relating to the applications of hybrids to PDT have been previously reported, most of them only focus on the designs of smart hybrids integrating multimodal imaging-guided multiple treatment modalities. Traditional PDT treatment has several limitations, such as inadequate PDT agents accumulating in cancer tissues, inferior PDT effect due to the devastating cancer hypoxia environment, relevant systemic toxicity in non-intelligent stimulation response treatment systems, and serious dependence of PDT on external light sources. Many strategies have been developed for overcoming these limitations, including improvement of cancer-homing ability by introducing active targeting groups, remodeling of the cancer hypoxia environment through oxygen regulators, intratumor release of ROS through activatable molecules, and replacement of laser light by X-rays or self-luminescence. This review aims to summarize the most recent advances in designing hybrids for improving the therapeutic efficacy of PDT.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Taixing Zhang
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | - Yong Hu
- Institute of Materials Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu 210093, China. and Shenzhen Research Institute of Nanjing University, Shenzhen, 518057, China
| |
Collapse
|
19
|
Xie S, Ai L, Cui C, Fu T, Cheng X, Qu F, Tan W. Functional Aptamer-Embedded Nanomaterials for Diagnostics and Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9542-9560. [PMID: 33595277 DOI: 10.1021/acsami.0c19562] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the past decades, various nanomaterials with unique properties have been explored for bioapplications. Meanwhile, aptamers, generated from the systematic evolution of ligands by exponential enrichment technology, are becoming an indispensable element in the design of functional nanomaterials because of their small size, high stability, and convenient modification, especially endowing nanomaterials with recognition capability to specific targets. Therefore, the incorporation of aptamers into nanomaterials offers an unprecedented opportunity in the research fields of diagnostics and therapeutics. Here, we focus on recent advances in aptamer-embedded nanomaterials for bioapplications. First, we briefly introduce the properties of nanomaterials that can be functionalized with aptamers. Then, the applications of aptamer-embedded nanomaterials in cellular analysis, imaging, targeted drug delivery, gene editing, and cancer diagnosis/therapy are discussed. Finally, we provide some perspectives on the challenges and opportunities that have arisen from this promising area.
Collapse
Affiliation(s)
- Sitao Xie
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Lili Ai
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, P. R. China
| | - Cheng Cui
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, P. R. China
| | - Ting Fu
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Xiangdong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Fengli Qu
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- College of Chemistry and Chemical, Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Weihong Tan
- The Cancer Hospital of the University of Chinese Academy of Sciences, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, P. R. China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
20
|
Zafar M, Ijaz M, Iqbal T. Efficient Au nanostructures for NIR-responsive controlled drug delivery systems. CHEMICAL PAPERS 2021. [DOI: 10.1007/s11696-020-01465-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Size-modulated optical property of gold nanorods for sensitive and colorimetric detection of thiourea in fruit juice. Talanta 2020; 225:121965. [PMID: 33592719 DOI: 10.1016/j.talanta.2020.121965] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/12/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
As an important sulfur compound, thiourea (TU) has caused great concern because of its wide application as well as its serious toxicity and hazard to the environment. Thus, it is necessary to develop a sensitive and selective method for TU analysis. In this work, gold nanorods (AuNRs) acted as an optical probe to realize the sensitive and colorimetric detection of TU. In HCl medium, Fe3+ at low concentration was difficult to oxide Au0 to form Au+ because of the high redox potential or the positive Gibbs free energy change. However, this process was possible when TU was present since the association constant between Au+ and TU is great enough to bind with TU to form a stable complex to further promote the etching of AuNRs, resulting in the lower aspect ratio of AuNRs with the blue shift and intensity decrease in extinction spectra, accompanied by the divisive colors of AuNRs solution or colorful dark-field light scattering imaging of single AuNR. The blue-shift of AuNRs longitudinal plasmon resonance absorption (LPRA) band was proportional to the concentration of TU in the range of 1-250 nM and the limit of detection (3σ/k) was as low as 0.4 nM. In addition, the colorimetric method was proven with high selectivity in the presence of potential interfering compounds, which was successfully applied to the detection of TU in fruit juice samples. This proposed colorimetric method provides a simple, sensitive yet selective measurement tool for TU sensing, which may offer new opportunities in the development of colorimetric sensors for food safety in the future.
Collapse
|
22
|
Liu JJ, Yan HH, Yuan D, Zhang Q, Li CM, Huang CZ, Wang J. The synergistic effect enhanced chemical etching of gold nanorods for the rapid and sensitive detection of biomarks. Talanta 2020; 219:121203. [DOI: 10.1016/j.talanta.2020.121203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
|
23
|
Nair RV, Nair LV, Govindachar DM, Santhakumar H, Nazeer SS, Rekha CR, Shenoy SJ, Periyasamy G, Jayasree RS. Luminescent Gold Nanorods To Enhance the Near‐Infrared Emission of a Photosensitizer for Targeted Cancer Imaging and Dual Therapy: Experimental and Theoretical Approach. Chemistry 2020; 26:2826-2836. [DOI: 10.1002/chem.201904952] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Resmi V. Nair
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
| | - Lakshmi V. Nair
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
- Present Address: School of Materials Science & Engineering National Institute of Technology Calicut, Kozhikode 673601 Kerala India
| | | | - Hema Santhakumar
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
| | - Shaiju S. Nazeer
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
- Present Address: Department of Chemistry University of Alabama at Birmingham Birmingham AL 35205 USA
| | | | - Sachin J. Shenoy
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
| | - Ganga Periyasamy
- Department of Chemistry Bangalore University Bangalore 560 056 India
| | - Ramapurath S. Jayasree
- Sree Chitra Tirunal Institute for Medical Sciences and Technology Thiruvananthapuram 695012 India
| |
Collapse
|
24
|
Fang Z, Wang X, Sun Y, Fan R, Liu Z, Guo R, Xie D. Sgc8 aptamer targeted glutathione-responsive nanoassemblies containing Ara-C prodrug for the treatment of acute lymphoblastic leukemia. NANOSCALE 2019; 11:23000-23012. [PMID: 31769777 DOI: 10.1039/c9nr07391d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cytarabine (Ara-C) is an essential medicine used in the clinical treatment of acute lymphoblastic leukemia. However, Ara-C suffers from high hydrophilicity, rapid plasma degradation and significant side effects. Thus, herein, to eliminate the limitations of Ara-C in the treatment of leukemia, Sgc8 aptamer targeting and glutathione (GSH)-responsive polymeric micelles (PCL-ss-Ara@Sgc8-BSA) were prepared. The prodrug was synthesized via covalent bond formation between acryloyl chloride-terminal PCL-ss-PCL and Ara-C, and surface decoration with Sgc8-bovine serum albumin (Sgc8-BSA). The obtained PCL-ss-Ara@Sgc8-BSA exhibited good GSH-responsive drug release behavior, obvious targetability and sufficient antitumor effect to acute lymphoblastic leukemia (ALL) cells (CCRF-CEM). A hemolysis test was further carried out to demonstrate that these polymeric micelles are safe to be administrated intravenously. Compared with free Ara-C, PCL-ss-Ara@Sgc8-BSA significantly enhanced tumor growth inhibition in mice bearing CCRF-CEM xenograft tumors, while causing little side effects, and improved the survival rate of CCRF-CEM tumor-bearing mice in vivo. Therefore, this new self-assembling small molecular prodrug equipped with Sgc8 targeting function is a potential treatment for the targeted therapy of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Zhigang Fang
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510010, China
| | - Xiaozhen Wang
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510010, China
| | - Yanling Sun
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510010, China
| | - Ruifang Fan
- Department of Prevention & Healthcare, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510010, China
| | - Zhong Liu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Deming Xie
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
25
|
Wang H, Rao H, Luo M, Xue X, Xue Z, Lu X. Noble metal nanoparticles growth-based colorimetric strategies: From monocolorimetric to multicolorimetric sensors. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.06.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
26
|
Yang Y, Chen M, Wu Y, Wang P, Zhao Y, Zhu W, Song Z, Zhang XB. Ultrasound assisted one-step synthesis of Au@Pt dendritic nanoparticles with enhanced NIR absorption for photothermal cancer therapy. RSC Adv 2019; 9:28541-28547. [PMID: 35529621 PMCID: PMC9071120 DOI: 10.1039/c9ra04286e] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022] Open
Abstract
Near-infrared (NIR) light-mediated non-invasive photothermal therapy (PTT) has attracted considerable attention for cancer treatment. Strong optical absorption located in the NIR region and high performance in converting light to heat should be emphasized for the development of ideal photothermal agents. In this report, Au@Pt bimetallic nanoparticles (Au@Pt NPs) with dendritic structure were synthesized through an ultrasound assisted one-step method in aqueous solution. The absorption of Au@Pt NPs at 808 nm was obviously enhanced compared to that of Au NPs and could be easily manipulated via the amount of Pt NPs. Au@Pt NPs exhibited excellent photostability with a photothermal conversion efficiency of 44.2%, which is significantly higher than those in most reported studies. Au@Pt NPs with thiol PEG functionalization presented improved cellular killing capacity upon NIR laser irradiation. Moreover, the potential application of Au@Pt NPs was also investigated in xenograft tumor mouse model. Overall, the remarkable therapeutic characteristics of PEGylated Au@Pt NPs provide them with great potential for future cancer treatment.
Collapse
Affiliation(s)
- Yue Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Mei Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Yajiao Wu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Peng Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Yan Zhao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Wenxiang Zhu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| | - Zhiling Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology Qingdao 266042 PR China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University Changsha 410082 PR China
| |
Collapse
|
27
|
Zhang L, Yang XQ, Wei JS, Li X, Wang H, Zhao YD. Intelligent gold nanostars for in vivo CT imaging and catalase-enhanced synergistic photodynamic & photothermal tumor therapy. Theranostics 2019; 9:5424-5442. [PMID: 31534494 PMCID: PMC6735389 DOI: 10.7150/thno.33015] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/06/2019] [Indexed: 12/23/2022] Open
Abstract
Photodynamic therapy (PDT) is a clinically approved and minimally invasive form of cancer treatment. However, due to hypoxia at the tumor site and phototoxicity to normal tissues, monotherapies using photosensitizers remain suboptimal. This study aimed to develop a highly selective controlled catalase-enhanced synergistic photodynamic and photothermal cancer therapy based on gold nanostars. Methods: Gold nanostars (GNS) with high thermal conversion efficiency were used as the core for photothermal therapy (PTT) and the shell consisted of the photosensitizer Ce6-loaded mesoporous silicon. The shell was modified with catalase (E), which catalyzes the conversion of hydrogen peroxide to oxygen at the tumor site, alleviating hypoxia and increasing the effect of the photodynamic treatment. Finally, a phospholipid derivative with c(RGDyK) was used as the targeting moiety and the nanoparticle-encapsulating material. Results: The nanoprobe exhibited good dispersion, high stability, and high photothermal conversion efficiency (~28%) for PTT as well as a photodynamic "on-off" effect on Ce6 encapsulated in mesoporous channels. The "release" of Ce6 was only triggered under photothermal stimulation in vivo. Due to its targeting ability, 72 h after injection of the probe, the tumor site in mice showed an observable CT response. The combined treatment using photothermal therapy (PTT) and catalase-enhanced photo-controlled PDT exerted a superior effect to PTT or PDT monotherapies. Conclusion: Our findings demonstrate that the use of this intelligent nanoprobe for CT-targeted image-guided treatment of tumors with integrated photothermal therapy (PTT) and catalase-enhanced controlled photodynamic therapy (PDT) may provide a novel approach for cancer theranostics.
Collapse
|
28
|
Yuan D, Liu JJ, Yan HH, Li CM, Huang CZ, Wang J. Label-free gold nanorods sensor array for colorimetric detection and discrimination of biothiols in human urine samples. Talanta 2019; 203:220-226. [PMID: 31202329 DOI: 10.1016/j.talanta.2019.05.076] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/01/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022]
Abstract
Biothiols play important roles in regulating redox balance in biological systems, but their discrimination is challengeable. In this work, a colorimetric nanosensing array for biothiols was established, which was composed of gold nanorods (AuNRs) and metal ions (Hg2+, Pb2+, Cu2+, Ag+). By employing label-free AuNRs as the colorimetric probe, and the color and spectral changes of AuNRs as the output signal, principal component analysis (PCA) was applied to processing the signal and generating a clustering map. Due to the different binding affinity between biothiols and metal ions, AuNRs exhibited a unique pattern to form a fingerprint-like colorimetric array, which was able to discriminate five biothiols by the naked eyes. This strategy combines PCA and sensor array to achieve rapid and accurate discrimination and detection of biothiols. In addition, the method shows the great potential in analysis of biothiols in human urine samples.
Collapse
Affiliation(s)
- Dan Yuan
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Jia Jun Liu
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Hui Hong Yan
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Chun Mei Li
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Cheng Zhi Huang
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Biomedical Analysis (Southwest University), Chongqing Science & Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400716, China.
| | - Jian Wang
- Key Laboratory on Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
29
|
Hlapisi N, Motaung TE, Linganiso LZ, Oluwafemi OS, Songca SP. Encapsulation of Gold Nanorods with Porphyrins for the Potential Treatment of Cancer and Bacterial Diseases: A Critical Review. Bioinorg Chem Appl 2019; 2019:7147128. [PMID: 31182957 PMCID: PMC6515112 DOI: 10.1155/2019/7147128] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/04/2019] [Indexed: 01/23/2023] Open
Abstract
Cancer and bacterial diseases have been the most incidental diseases to date. According to the World Health Report 2018, at least every family is affected by cancer around the world. In 2012, 14.1 million people were affected by cancer, and that figure is bound to increase to 21.6 million in 2030. Medicine therefore sorts out ways of treatment using conventional methods which have been proven to have many side effects. Researchers developed photothermal and photodynamic methods to treat both cancer and bacterial diseases. These methods pose fewer effects on the biological systems but still no perfect method has been synthesized. The review serves to explore porphyrin and gold nanorods to be used in the treatment of cancer and bacterial diseases: porphyrins as photosensitizers and gold nanorods as delivery agents. In addition, the review delves into ways of incorporating photothermal and photodynamic therapy aimed at producing a less toxic, more efficacious, and specific compound for the treatment.
Collapse
Affiliation(s)
- Nthabeleng Hlapisi
- Department of Chemistry, University of Zululand, X1001, KwaDlangezwa, KwaZulu-Natal, South Africa
| | - Tshwafo E. Motaung
- Department of Chemistry, University of Zululand, X1001, KwaDlangezwa, KwaZulu-Natal, South Africa
| | - Linda Z. Linganiso
- Department of Chemistry, University of Zululand, X1001, KwaDlangezwa, KwaZulu-Natal, South Africa
| | - Oluwatobi S. Oluwafemi
- Department of Applied Chemistry, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg 2028, South Africa
- Centre for Nanomaterials Science Research, University of Johannesburg, Johannesburg, South Africa
| | - Sandile P. Songca
- Department of Chemistry, University of Kwazulu Natal, Kwazulu Natal, South Africa
| |
Collapse
|
30
|
Munzar JD, Ng A, Juncker D. Duplexed aptamers: history, design, theory, and application to biosensing. Chem Soc Rev 2019; 48:1390-1419. [PMID: 30707214 DOI: 10.1039/c8cs00880a] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nucleic acid aptamers are single stranded DNA or RNA sequences that specifically bind a cognate ligand. In addition to their widespread use as stand-alone affinity binding reagents in analytical chemistry, aptamers have been engineered into a variety of ligand-specific biosensors, termed aptasensors. One of the most common aptasensor formats is the duplexed aptamer (DA). As defined herein, DAs are aptasensors containing two nucleic acid elements coupled via Watson-Crick base pairing: (i) an aptamer sequence, which serves as a ligand-specific receptor, and (ii) an aptamer-complementary element (ACE), such as a short DNA oligonucleotide, which is designed to hybridize to the aptamer. The ACE competes with ligand binding, such that DAs generate a signal upon ligand-dependent ACE-aptamer dehybridization. DAs possess intrinsic advantages over other aptasensor designs. For example, DA biosensing designs generalize across DNA and RNA aptamers, DAs are compatible with many readout methods, and DAs are inherently tunable on the basis of nucleic acid hybridization. However, despite their utility and popularity, DAs have not been well defined in the literature, leading to confusion over the differences between DAs and other aptasensor formats. In this review, we introduce a framework for DAs based on ACEs, and use this framework to distinguish DAs from other aptasensor formats and to categorize cis- and trans-DA designs. We then explore the ligand binding dynamics and chemical properties that underpin DA systems, which fall under conformational selection and induced fit models, and which mirror classical SN1 and SN2 models of nucleophilic substitution reactions. We further review a variety of in vitro and in vivo applications of DAs in the chemical and biological sciences, including riboswitches and riboregulators. Finally, we present future directions of DAs as ligand-responsive nucleic acids. Owing to their tractability, versatility and ease of engineering, DA biosensors bear a great potential for the development of new applications and technologies in fields ranging from analytical chemistry and mechanistic modeling to medicine and synthetic biology.
Collapse
Affiliation(s)
- Jeffrey D Munzar
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
31
|
Yang S, Li H, Xu L, Deng Z, Han W, Liu Y, Jiang W, Zu Y. Oligonucleotide Aptamer-Mediated Precision Therapy of Hematological Malignancies. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:164-175. [PMID: 30292138 PMCID: PMC6172475 DOI: 10.1016/j.omtn.2018.08.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/31/2018] [Accepted: 08/31/2018] [Indexed: 01/01/2023]
Abstract
Precision medicine has recently emerged as a promising strategy for cancer therapy because it not only specifically targets cancer cells but it also does not have adverse effects on normal cells. Oligonucleotide aptamers are a class of small molecule ligands that can specifically bind to their targets on cell surfaces with high affinity. Aptamers have great potential in precision cancer therapy due to their unique physical, chemical, and biological properties. Therefore, aptamer technology has been widely investigated for biomedical and clinical applications. This review focuses on the potential applications of aptamer technology as a new tool for precision treatment of hematological malignancies, including leukemia, lymphoma, and multiple myeloma.
Collapse
Affiliation(s)
- Shuanghui Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Huan Li
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA; Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ling Xu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA; Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Zhenhan Deng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wei Han
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Yanting Liu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Wenqi Jiang
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Chien YH, Chan KK, Anderson T, Kong KV, Ng BK, Yong KT. Advanced Near-Infrared Light-Responsive Nanomaterials as Therapeutic Platforms for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800090] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yi-Hsin Chien
- School of Electrical and Electronic Engineering; Nanyang Technological University; Singapore 639798
- Department of Materials Science and Engineering; Feng Chia University; Taichung 40724 Taiwan
| | - Kok Ken Chan
- School of Electrical and Electronic Engineering; Nanyang Technological University; Singapore 639798
| | - Tommy Anderson
- School of Electrical and Electronic Engineering; Nanyang Technological University; Singapore 639798
| | - Kien Voon Kong
- Department of Chemistry; National Taiwan University; Taipei 10617 Taiwan
| | - Beng Koon Ng
- School of Electrical and Electronic Engineering; Nanyang Technological University; Singapore 639798
| | - Ken-Tye Yong
- School of Electrical and Electronic Engineering; Nanyang Technological University; Singapore 639798
| |
Collapse
|
33
|
Kruger CA, Abrahamse H. Utilisation of Targeted Nanoparticle Photosensitiser Drug Delivery Systems for the Enhancement of Photodynamic Therapy. Molecules 2018; 23:E2628. [PMID: 30322132 PMCID: PMC6222717 DOI: 10.3390/molecules23102628] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/25/2022] Open
Abstract
The cancer incidence world-wide has caused an increase in the demand for effective forms of treatment. One unconventional form of treatment for cancer is photodynamic therapy (PDT). PDT has 3 fundamental factors, namely a photosensitiser (PS) drug, light and oxygen. When a PS drug is administered to a patient, it can either passively or actively accumulate within a tumour site and once exposed to a specific wavelength of light, it is excited to produce reactive oxygen species (ROS), resulting in tumour destruction. However, the efficacy of ROS generation for tumour damage is highly dependent on the uptake of the PS in tumour cells. Thus, PS selective/targeted uptake and delivery in tumour cells is a crucial factor in PDT cancer drug absorption studies. Generally, within non-targeted drug delivery mechanisms, only minor amounts of PS are able to passively accumulate in tumour sites (due to the enhanced permeability and retention (EPR) effect) and the remainder distributes into healthy tissues, causing unwanted side effects and poor treatment prognosis. Thus, to improve the efficacy of PDT cancer treatment, research is currently focused on the development of specific receptor-based PS-nanocarrier platform drugs, which promote the active uptake and absorption of PS drugs in tumour sites only, avoiding unwanted side effects, as well as treatment enhancement. Therefore, the aim of this review paper is to focus on current actively targeted or passively delivered PS nanoparticle drug delivery systems, that have been previously investigated for the PDT treatment of cancer and so to deduce their overall efficacy and recent advancements.
Collapse
Affiliation(s)
- Cherie Ann Kruger
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, Doornfontein 2001, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, Doornfontein 2001, South Africa.
| |
Collapse
|
34
|
Wang S, Guo F, Ji Y, Yu M, Wang J, Li N. Dual-Mode Imaging Guided Multifunctional Theranosomes with Mitochondria Targeting for Photothermally Controlled and Enhanced Photodynamic Therapy in Vitro and in Vivo. Mol Pharm 2018; 15:3318-3331. [DOI: 10.1021/acs.molpharmaceut.8b00351] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Siyu Wang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, PR China
| | - Fang Guo
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, PR China
| | - Yanhui Ji
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, 300052 Tianjin, PR China
| | - Meng Yu
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, PR China
| | - Jinping Wang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, PR China
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, PR China
| |
Collapse
|
35
|
Zou X, Yang F, Sun X, Qin M, Zhao Y, Zhang Z. Functionalized Nano-adsorbent for Affinity Separation of Proteins. NANOSCALE RESEARCH LETTERS 2018; 13:165. [PMID: 29846826 PMCID: PMC5976561 DOI: 10.1186/s11671-018-2531-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 04/16/2018] [Indexed: 05/07/2023]
Abstract
Thiol-functionalized silica nanospheres (SiO2-SH NSs) with an average diameter of 460 nm were synthesized through a hydrothermal route. Subsequently, the prepared SiO2-SH NSs were modified by SnO2 quantum dots to afford SnO2/SiO2 composite NSs possessing obvious fluorescence, which could be used to trace the target protein. The SnO2/SiO2 NSs were further modified by reduced glutathione (GSH) to obtain SnO2/SiO2-GSH NSs, which can specifically separate glutathione S-transferase-tagged (GST-tagged) protein. Moreover, the peroxidase activity of glutathione peroxidase 3 (GPX3) separated from SnO2/SiO2-GSH NSs in vitro was evaluated. Results show that the prepared SnO2/SiO2-GSH NSs exhibit negligible nonspecific adsorption, high concentration of protein binding (7.4 mg/g), and good reused properties. In the meantime, the GST-tagged GPX3 separated by these NSs can retain its redox state and peroxidase activity. Therefore, the prepared SnO2/SiO2-GSH NSs might find promising application in the rapid separation and purification of GST-tagged proteins.
Collapse
Affiliation(s)
- Xueyan Zou
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004 China
- National & Local Joint Engineering Research Center for Applied Technology of Hybrid Nanomaterials, Henan University, Kaifeng, 475004 China
- Collaborative Innovation Center of Nano Functional Materials and Applications of Henan Province, Henan University, Kaifeng, 475004 China
| | - Fengbo Yang
- Institute of Plant Stress Biology-State Key Laboratory of Cotton Biology, Henan University, Kaifeng, 475004 China
| | - Xin Sun
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004 China
| | - Mingming Qin
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004 China
| | - Yanbao Zhao
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004 China
- National & Local Joint Engineering Research Center for Applied Technology of Hybrid Nanomaterials, Henan University, Kaifeng, 475004 China
- Collaborative Innovation Center of Nano Functional Materials and Applications of Henan Province, Henan University, Kaifeng, 475004 China
| | - Zhijun Zhang
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004 China
- National & Local Joint Engineering Research Center for Applied Technology of Hybrid Nanomaterials, Henan University, Kaifeng, 475004 China
- Collaborative Innovation Center of Nano Functional Materials and Applications of Henan Province, Henan University, Kaifeng, 475004 China
| |
Collapse
|
36
|
Xin J, Wang S, Wang B, Wang J, Wang J, Zhang L, Xin B, Shen L, Zhang Z, Yao C. AlPcS 4-PDT for gastric cancer therapy using gold nanorod, cationic liposome, and Pluronic ® F127 nanomicellar drug carriers. Int J Nanomedicine 2018; 13:2017-2036. [PMID: 29670347 PMCID: PMC5894760 DOI: 10.2147/ijn.s154054] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose As a promising photodynamic therapy (PDT) agent, Al(III) phthalocyanine chloride tetrasulfonic acid (AlPcS4) provides deep penetration into tissue, high quantum yields, good photostability, and low photobleaching. However, its low delivery efficiency and high binding affinity to serum albumin cause its low penetration into cancer cells, further limiting its PDT effect on gastric cancer. In order to improve AlPcS4/PDT effect, the AlPcS4 delivery sys tems with different drug carriers were synthesized and investigated. Materials and methods Gold nanorods, cationic liposomes, and Pluronic® F127 nanomicellars were used to formulate the AlPcS4 delivery systems. The anticancer effect was evaluated by CCK-8 assay and colony formation assay. The delivery efficiency of AlPcS4 and the binding affinity to serum proteins were determined by fluorescence intensity assay. The apoptosis and necrosis ability, reactive oxygen species and singlet oxygen generation, mitochondrial transmembrane potential and ([Ca2+]i) concentration were further measured to evaluate the mechanism of cell death. Results The series of synthesized AlPcS4 delivery systems with different drug carriers improve the limited PDT effect in varying degrees. In contrast, AlPcS4 complex with gold nanorods has significant anticancer effects because gold nanorods are not only suitable for AlPcS4 delivery, but also exhibit enhanced singlet oxygen generation effect and photothermal effect to induce cell death directly. Moreover, AlPcS4 complex with cationic liposomes shows the potent inhibition effect because of its optimal AlPcS4 delivery efficiency and ability to block serum albumin. In addition, AlPcS4 complex with Pluronic F127 exhibits inferior PDT effect but presents lower cytotoxicity, slower dissociation rate, and longer retention time of incorporated drugs; thus, F127–AlPcS4 is used for prolonged gastric cancer therapy. Conclusion The described AlPcS4 drug delivery systems provide promising agents for gastric cancer therapy.
Collapse
Affiliation(s)
- Jing Xin
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sijia Wang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bing Wang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiazhuang Wang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Wang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Luwei Zhang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Xin
- School of Innovation and Entrepreneurship, Xi'an Fan Yi University, Xi'an, Shaanxi, China
| | - Lijian Shen
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenxi Zhang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Cuiping Yao
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Biomedical Analytical Technology and Instrumentation, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
37
|
Chen S, Fan J, Qiu W, Liu F, Yan G, Zeng X, Zhang X. A cellular/intranuclear dual-targeting nanoplatform based on gold nanostar for accurate tumor photothermal therapy. J Mater Chem B 2018; 6:1543-1551. [DOI: 10.1039/c8tb00087e] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A versatile GNS-NLS@HA nanoplatform was constructed for tumor cellular/intranuclear dual-targeting photothermal therapy. It displayed excellent tumor inhibition efficiency as well as anti-metastasis ability in vivo.
Collapse
Affiliation(s)
- Si Chen
- School of Material Science and Engineering
- Wuhan Institute of Technology
- Wuhan 430205
- P. R. China
| | - Jinxuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Wenxiu Qiu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Fan Liu
- School of Material Science and Engineering
- Wuhan Institute of Technology
- Wuhan 430205
- P. R. China
| | - Guoping Yan
- School of Material Science and Engineering
- Wuhan Institute of Technology
- Wuhan 430205
- P. R. China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| | - Xianzheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry
- Wuhan University
- Wuhan 430072
- P. R. China
| |
Collapse
|
38
|
Zhao N, Zeng Z, Zu Y. Self-Assembled Aptamer-Nanomedicine for Targeted Chemotherapy and Gene Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:10.1002/smll.201702103. [PMID: 29205808 PMCID: PMC5857619 DOI: 10.1002/smll.201702103] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/25/2017] [Indexed: 05/20/2023]
Abstract
Chemotherapy is the mainstream treatment of anaplastic large cell lymphoma (ALCL). However, chemotherapy can cause severe adverse effects in patients because it is not ALCL-specific. In this study, a multifunctional aptamer-nanomedicine (Apt-NMed) achieving targeted chemotherapy and gene therapy of ALCL is developed. Apt-NMed is formulated by self-assembly of synthetic oligonucleotides containing CD30-specific aptamer and anaplastic lymphoma kinase (ALK)-specific siRNA followed by self-loading of the chemotherapeutic drug doxorubicin (DOX). Apt-NMed exhibits a well-defined nanostructure (diameter 59 mm) and stability in human serum. Under aptamer guidance, Apt-NMed specifically binds and internalizes targeted ALCL cells. Intracellular delivery of Apt-NMed triggers rapid DOX release for targeted ALCL chemotherapy and intracellular delivery of the ALK-specific siRNA induced ALK oncogene silencing, resulting in combined therapeutic effects. Animal model studies reveal that upon systemic administration, Apt-NMed specifically targets and selectively accumulates in ALCL tumor site, but does not react with off-target tumors in the same xenograft mouse. Importantly, Apt-NMed not only induces significantly higher inhibition in ALCL tumor growth, but also causes fewer or no side effects in treated mice compared to free DOX. Moreover, Apt-NMed treatment markedly improves the survival rate of treated mice, opening a new avenue for precision treatment of ALCL.
Collapse
MESH Headings
- Animals
- Aptamers, Nucleotide/chemistry
- Doxorubicin/chemistry
- Doxorubicin/therapeutic use
- Genetic Therapy/methods
- Humans
- Lymphoma, Large-Cell, Anaplastic/drug therapy
- Lymphoma, Large-Cell, Anaplastic/mortality
- Lymphoma, Large-Cell, Anaplastic/therapy
- Mice
- Mice, SCID
- Microscopy, Electron, Scanning
- Microscopy, Fluorescence
- Nanomedicine/methods
- Nanostructures/chemistry
- RNA, Small Interfering/genetics
- RNA, Small Interfering/physiology
- Survival Rate
- U937 Cells
Collapse
Affiliation(s)
| | | | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin St., Houston, TX 77030, USA
| |
Collapse
|
39
|
Lim DJ, Park H. Near-infrared light for on-demand drug delivery. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 29:750-761. [PMID: 29082832 DOI: 10.1080/09205063.2017.1398994] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There are currently many basic technologies for the controlled release of therapeutic molecules for the treatment of chronic pathologies such as arthritis, asthma, and diabetes. Examples of such technologies include selectively dissolvable capsules and tablets that are designed to respond to specific stimuli - such as pH, temperature, or specific enzymes - in a time-specific fashion. However, because of the biological variations between different individuals, which contribute to differences in the environments of therapeutic target locations, these technologies are not fully controllable. In the pursuit of drug-release technologies that are fully controllable, many approaches have been examined. One such approach involves the utilization of various light-sensitive molecules that are designed to release therapeutic agents when stimulated by light of specific wavelengths. Potential light sources that have been explored for this approach include ultraviolet (UV) and near-infrared (NIR) light. UV light, which exists in the range of 10-400 nm, is easily to utilize, and many chemicals and particles can be stimulated with light in this spectrum. Unfortunately, when used extensively - as would be the case for chronic pathologies - UV light can cause cellular damage at the molecular level, potentially leading to skin cancer. A viable alternative to UV light is NIR light, which offers deeper transdermal penetration and does not have many known adverse long-term side effects. Therefore, the purpose of this review is to investigate the use of NIR light and the associated therapeutic molecules for the controlled release of therapeutic agents in the potential treatment of chronic pathologies.
Collapse
Affiliation(s)
- Dong-Jin Lim
- a Department of Otolaryngology Head & Neck Surgery , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Hansoo Park
- b School of Integrative Engineering , Chung-Ang University , Seoul , Republic of Korea
| |
Collapse
|
40
|
Fluorescence guided photothermal/photodynamic ablation of tumours using pH-responsive chlorin e6-conjugated gold nanorods. Colloids Surf B Biointerfaces 2017; 160:345-354. [PMID: 28961542 DOI: 10.1016/j.colsurfb.2017.09.045] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/16/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023]
Abstract
Photothermal/photodynamic therapies (PTT/PDT) have been widely accepted as non-invasive therapeutic modalities to erase tumours. However, both therapies face the problem of precisely locating tumours and reducing their side effects. Herein, chlorin e6 conjugated gold nanorod, (Ce6-PEG-AuNR), a type of gold nanorod-photosensitizer conjugate, is designed as a kind of nano-therapeutic agent to simultaneously realize combined PTT/PDT. Compared to free Ce6, the fluorescence of Ce6 adhered to the conjugate is effectively quenched by the longitudinal surface plasmon resonance (LSPR) of in the Ce6-PEG-AuNR. However, the specific fluorescence of Ce6 can be recovered in tumour tissue when Ce6 is separated from the conjugate owing to the cleavage of hydrazone bond between Ce6 and PEG caused by intracellular acidic conditions in tumour tissue. Based on this effect, we can precisely locate tumours and further kill cancer cells by combined PTT/PDT. In addition, the combined therapy (PTT/PDT) function is more efficient in cancer treatment than that of PTT or PDT alone. Therefore, Ce6-PEG-AuNR can serve as a promising dual-modal phototherapeutic agent as well as a tumour-sensitive fluorescent probe to diagnose and treat cancer.
Collapse
|
41
|
Wu D, Wang L, Li W, Xu X, Jiang W. DNA nanostructure-based drug delivery nanosystems in cancer therapy. Int J Pharm 2017; 533:169-178. [PMID: 28923770 DOI: 10.1016/j.ijpharm.2017.09.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 01/04/2023]
Abstract
DNA as a novel biomaterial can be used to fabricate different kinds of DNA nanostructures based on its principle of GC/AT complementary base pairing. Studies have shown that DNA nanostructure is a nice drug carrier to overcome big obstacles existing in cancer therapy such as systemic toxicity and unsatisfied drug efficacy. Thus, different types of DNA nanostructure-based drug delivery nanosystems have been designed in cancer therapy. To improve treating efficacy, they are also developed into more functional drug delivery nanosystems. In recent years, some important progresses have been made. The objective of this review is to make a retrospect and summary about these different kinds of DNA nanostructure-based drug delivery nanosystems and their latest progresses: (1) active targeting; (2) mutidrug co-delivery; (3) construction of stimuli-responsive/intelligent nanosystems.
Collapse
Affiliation(s)
- Dandan Wu
- Key Laboratory of Natural Products Chemical Biology, Ministry of Education, School of Pharmacy, Shandong University, Jinan, 250012, PR China
| | - Lei Wang
- Key Laboratory of Natural Products Chemical Biology, Ministry of Education, School of Pharmacy, Shandong University, Jinan, 250012, PR China
| | - Wei Li
- Key Laboratory of Natural Products Chemical Biology, Ministry of Education, School of Pharmacy, Shandong University, Jinan, 250012, PR China
| | - Xiaowen Xu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, PR China
| | - Wei Jiang
- Key Laboratory of Natural Products Chemical Biology, Ministry of Education, School of Pharmacy, Shandong University, Jinan, 250012, PR China; School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, PR China.
| |
Collapse
|
42
|
Wang S, Li C, Meng Y, Qian M, Jiang H, Du Y, Huang R, Wang Y. MemHsp70 Receptor-mediated Multifunctional Ordered Mesoporous Carbon Nanospheres for Photoacoustic Imaging-Guided Synergistic Targeting Trimodal Therapy. ACS Biomater Sci Eng 2017; 3:1702-1709. [DOI: 10.1021/acsbiomaterials.7b00326] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Shanshan Wang
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Chengyi Li
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Ying Meng
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Min Qian
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Huiling Jiang
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yilin Du
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Rongqin Huang
- Department of Pharmaceutics, School of
Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yi Wang
- Center for Advanced
Low-dimension Materials, Donghua University, Shanghai 201620, China
- Center of Analysis and Measurement, Fudan University, Shanghai 200433, China
| |
Collapse
|
43
|
Yang X, Xue X, Luo Y, Lin TY, Zhang H, Lac D, Xiao K, He Y, Jia B, Lam KS, Li Y. Sub-100nm, long tumor retention SN-38-loaded photonic micelles for tri-modal cancer therapy. J Control Release 2017; 261:297-306. [PMID: 28700898 DOI: 10.1016/j.jconrel.2017.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/26/2017] [Accepted: 07/07/2017] [Indexed: 02/05/2023]
Abstract
The tumor penetration and accumulation of nanoparticle-based drug delivery systems are highly dependent on the particle size. Nanomedicines in the sub-100nm range have been suggested by previous studies to have superior antitumor efficacy on various solid tumors. SN-38 is a very important and highly potent drug for several cancers including colon cancer. However, due to the ultra-flat aromatic structure of SN-38, it is typically very difficult to produce sub-100nm, SN-38-encapsulated nanoparticles without modification of the chemical structure. Here, we report on the successful production of 20-30nm, SN-38-encapsulated photonic micelles for effectively trimodal cancer therapy. Taking advantages of the supramolecular "π-π" stacking and hydrophobicity interaction between SN-38, and a unique class of photonic nanoporphyrin micelles (NPM), the extremely hydrophobic SN-38 was successfully encapsulated into NPM with significantly increased water solubility (up to 500 times). At equivalent dose of drug, photosensitizer and light irradiation, combination therapy with SN-38-encapsulated nanoporphyrin micelles (SN-NPM) enhanced the in vitro antitumor activity by 78 and 350 times over single treatment with SN-38 and phototherapy alone, respectively. Due to the relatively small size, SN-NPM possessed superior long tumor retention time (>5days) and much higher accumulation in tumors than in normal organs, as shown by near-infrared fluorescence (NIRF) imaging. Furthermore, the trimodal therapy (photothermal-, photodynamic- and chemo-therapy) with SN-NPM demonstrated dramatically enhanced in vivo antitumor efficacy over single treatment on nude mice bearing HT-29 colon cancer xenograft. Therefore, these sub-100nm, SN-38-encapsulated photonic micelles show great promise for multimodal cancer therapy.
Collapse
Affiliation(s)
- Xixiao Yang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA; Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China
| | - Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Yan Luo
- Department of Oncology, 153 Central Hospital, Zhengzhou 450042, PR China
| | - Tzu-Yin Lin
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Hongyong Zhang
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Diana Lac
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yixuan He
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Bei Jia
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA; Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
44
|
Abstract
Leukemia is a cancer of blood cells and bone marrow, leading to death in many patients mainly in children. Over the last several years, aptamers generated by SELEX (Systematic evolution of ligands by exponential enrichment) method, have quickly become a new class of targeting ligands for drug delivery applications and recently have been widely exploited in different biomedical applications, due to several potent properties such as high binding affinity and selectivity, low or no immunogenicity and toxicity, low cost and thermal stability. In this review, we presented in details about aptamers involved in targeting, and treatment of leukemia. Moreover, some analytical approaches such as electrochemical and optical aptasensors were introduced for detection and diagnosis of leukemia. Finally, we discussed about the directions and challenges of aptamer application in this field.
Collapse
|
45
|
Yeo ELL, Cheah JUJ, Lim BY, Thong PSP, Soo KC, Kah JCY. Protein Corona around Gold Nanorods as a Drug Carrier for Multimodal Cancer Therapy. ACS Biomater Sci Eng 2017; 3:1039-1050. [PMID: 33429578 DOI: 10.1021/acsbiomaterials.7b00231] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A single nanodevice based on gold nanorods (NRs) coloaded with a photosensitizer, Chlorin e6 (Ce6), and a chemotherapeutic, Doxorubicin (Dox), on its endogenously formed human serum (HS) protein corona, i.e., NR-HS-Ce6-Dox was developed with the aim of performing multimodal cancer therapy: photodynamic (PDT), photothermal (PTT) and chemotherapy (CTX) simultaneously upon irradiation with a single 665 nm laser. Here, the excitation of NRs and Ce6 resulted in photothermal ablation (PTT), and production of reactive oxygen species (ROS) to kill Cal 27 oral squamous cell carcinoma (OSCC) cells by oxidative stress (PDT) respectively, while the laser-triggered release of Dox intercalated into the DNA of cancer cells to result in DNA damage and cell death (CTX). High laser-triggered Dox release efficiency of 71.5% and strong plasmonic enhancement of ROS production by Ce6 (4.8-fold increase compared to free Ce6) was observed. Uptake of both Ce6 and Dox by Cal 27 cells was greatly enhanced, with 3.3 and 52 times higher intracellular Dox and Ce6 fluorescence observed, respectively, 6 h after dosing with NR-HS-Ce6-Dox compared to free drugs. The simultaneous trimodal therapy achieved a near complete eradication of cancer cells (98.7% cell death) with an extremely low dose of 15 pM NR-HS-Ce6-Dox loaded with just 1.26 nM Ce6 and 12.5 nM Dox due to strong synergistic enhancement in cancer cell kill compared to individual therapies performed separately. No dark toxicities were observed. These drug concentrations were far lower than any previously reported in vitro, thus eliminating any potential systemic toxicity of these agents.
Collapse
Affiliation(s)
- Eugenia Li Ling Yeo
- Department of Biomedical Engineering, National University of Singapore 4 Engineering Drive 3, E4-04-08, Singapore 117583
| | - Joshua U-Jin Cheah
- NUS Graduate School for Integrative Sciences and Engineering Centre for Life Sciences (CeLS), National University of Singapore, #05-01, 28 Medical Drive, Singapore 117456
| | - Bing Yi Lim
- Department of Biomedical Engineering, National University of Singapore 4 Engineering Drive 3, E4-04-08, Singapore 117583
| | - Patricia Soo Ping Thong
- Division of Medical Sciences, National Cancer Centre Singapore 11 Hospital Drive, Singapore 169610
| | - Khee Chee Soo
- Division of Medical Sciences, National Cancer Centre Singapore 11 Hospital Drive, Singapore 169610
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore 4 Engineering Drive 3, E4-04-08, Singapore 117583.,NUS Graduate School for Integrative Sciences and Engineering Centre for Life Sciences (CeLS), National University of Singapore, #05-01, 28 Medical Drive, Singapore 117456
| |
Collapse
|
46
|
Sun Q, You Q, Pang X, Tan X, Wang J, Liu L, Guo F, Tan F, Li N. A photoresponsive and rod-shape nanocarrier: Single wavelength of light triggered photothermal and photodynamic therapy based on AuNRs-capped & Ce6-doped mesoporous silica nanorods. Biomaterials 2017; 122:188-200. [DOI: 10.1016/j.biomaterials.2017.01.021] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 01/05/2017] [Accepted: 01/16/2017] [Indexed: 01/21/2023]
|
47
|
Zhang HZ, Li RS, Gao PF, Wang N, Lei G, Huang CZ, Wang J. Real-time dark-field light scattering imaging to monitor the coupling reaction with gold nanorods as an optical probe. NANOSCALE 2017; 9:3568-3575. [PMID: 28244517 DOI: 10.1039/c6nr09453h] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Gold nanorods (GNRs) have opened up promising applications based on their reshaping, due to the fact that a tiny change in shape or size could directly lead to optical changes. Herein, we report chemical reshaping of GNRs induced by the coupling reaction between Au, ferric chloride and thiourea. In the coupling reaction, Fe3+ oxidizes the GNRs to yield Au(i), which complexes with the thiourea ligand, lowering the Gibbs free energy of the gold species and promoting the reaction equilibrium to enable the chemical reshaping of the GNRs. This coupling reaction process was monitored using a light-scattering dark-field microscopy (DFM) imaging technique and scanning electron microscopy (SEM). The light scattering underwent a colour change from bright red to yellow and finally to green, and the GNRs underwent a morphological change from rod-shaped to fusiform and finally to spherical, which is somewhat different from the results of other chemical etching processes of GNRs. It is believed that the coupling reaction induced chemical reshaping of GNRs not only provides an alternative way to monitor the coupling reaction, but also offers a facile way to obtain a desirable GNR morphology, which is important for the preparation of fusiform nanostructures.
Collapse
Affiliation(s)
- Hong Zhi Zhang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| | - Rong Sheng Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| | - Peng Fei Gao
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| | - Ni Wang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| | - Gang Lei
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| | - Cheng Zhi Huang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China. and Chongqing Key Laboratory of Biomedical Analysis (Southwest University), Chongqing Science & Technology Commission, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Jian Wang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
48
|
Su L, Hu S, Zhang L, Wang Z, Gao W, Yuan J, Liu M. A Fast and Efficient Replacement of CTAB with MUA on the Surface of Gold Nanorods Assisted by a Water-Immiscible Ionic Liquid. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602809. [PMID: 28067980 DOI: 10.1002/smll.201602809] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/07/2016] [Indexed: 06/06/2023]
Abstract
The synthesis and surface modification of gold nanorods (GNRs) is one of the most important and basic issues in nanoscience. Most of the widely investigated GNRs are coated with a cetyltrimethylammonium bromide(CTAB) bilayer. Here, a highly efficient method is proposed to replace CTAB from the surface of GNRs with a bifunctional 11-mercaptoundecanoic acid in order to decrease the possible toxicity caused by CTAB. This ligand exchange is achieved in a biphasic mixture of an aqueous solution and a water-immiscible ionic liquid (IL), [BMIM][Tf2 N]. That is, by mixing IL, mercaptoundecanoic acid (MUA)/IL (200 × 10-3 m) and a concentrated aqueous solution of GNRs together, followed by vortex stirring for 90 s, CTAB-capped GNRs with varying aspect ratios can be turned into corresponding MUA-capped GNRs with the same aspect ratio. Furthermore, the formed MUA-capped GNRs can be obtained in a large quantity and stored as powders for easy use. The MUA-capped GNRs with improved biocompatibility and colloidal stability are well suited for further biological functionalization and potential applications. This IL-assisted ligand exchange can reverse the surface charge, enhance the stability of GNRs, and suppress its cytotoxicity.
Collapse
Affiliation(s)
- Linjia Su
- Department of Chemistry, Capital Normal University, Beijing, 100048, P. R. China
| | - Song Hu
- Department of Chemistry, Capital Normal University, Beijing, 100048, P. R. China
| | - Li Zhang
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Colloid, Interface, Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhuoran Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, P. R. China
| | - Weiping Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, P. R. China
| | - Jing Yuan
- Department of Chemistry, Capital Normal University, Beijing, 100048, P. R. China
| | - Minghua Liu
- Beijing National Laboratory for Molecular Science, CAS Key Laboratory of Colloid, Interface, Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
49
|
Leitner M, Poturnayova A, Lamprecht C, Weich S, Snejdarkova M, Karpisova I, Hianik T, Ebner A. Characterization of the specific interaction between the DNA aptamer sgc8c and protein tyrosine kinase-7 receptors at the surface of T-cells by biosensing AFM. Anal Bioanal Chem 2017; 409:2767-2776. [PMID: 28229174 PMCID: PMC5366180 DOI: 10.1007/s00216-017-0238-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/18/2017] [Accepted: 01/31/2017] [Indexed: 01/10/2023]
Abstract
We studied the interaction of the specific DNA aptamer sgc8c immobilized at the AFM tip with its corresponding receptor, the protein tyrosine kinase-7 (PTK7) embedded in the membrane of acute lymphoblastic leukemia (ALL) cells (Jurkat T-cells). Performing single molecule force spectroscopy (SMFS) experiments, we showed that the aptamer sgc8c bound with high probability (38.3 ± 7.48%) and high specificity to PTK7, as demonstrated by receptor blocking experiments and through comparison with the binding behavior of a nonspecific aptamer. The determined kinetic off-rate (koff = 5.16 s−1) indicates low dissociation of the sgc8c–PTK7 complex. In addition to the pulling force experiments, simultaneous topography and recognition imaging (TREC) experiments using AFM tips functionalized with sgc8c aptamers were realized on the outer regions surface of surface-immobilized Jurkat cells for the first time. This allowed determination of the distribution of PTK7 without any labeling and at near physiological conditions. As a result, we could show a homogeneous distribution of PTK7 molecules on the outer regions of ALL cells with a surface density of 325 ± 12 PTK7 receptors (or small receptor clusters) per μm2. The specific interaction of the DNA aptamer sgc8c and protein tyrosine kinase-7 (PTK7) on acute lymphoblastic leukemia (ALL) cells was characterized. AFM based single molecule force spectroscopy (SMFS) yielded a kinetic off-rate of 5.16 s−1 of the complex. Simultaneous topography and recognition imaging (TREC) revealed a PTK7 density of 325 ± 12 molecules or clusters per μm2 in the cell membrane ![]()
Collapse
Affiliation(s)
- Michael Leitner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Alexandra Poturnayova
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia.,Institute of Biochemistry and Animal Genetics, Slovak Academy of Sciences, Moyzesova 61, 900 28, Ivanka pri Dunaji, Slovakia
| | - Constanze Lamprecht
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Sabine Weich
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria
| | - Maja Snejdarkova
- Institute of Biochemistry and Animal Genetics, Slovak Academy of Sciences, Moyzesova 61, 900 28, Ivanka pri Dunaji, Slovakia
| | - Ivana Karpisova
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia
| | - Tibor Hianik
- Faculty of Mathematics, Physics, and Informatics, Comenius University, Mlynska dolina F1, 842 48, Bratislava, Slovakia
| | - Andreas Ebner
- Institute of Biophysics, Johannes Kepler University Linz, Gruberstrasse 40, 4020, Linz, Austria.
| |
Collapse
|
50
|
Yi Y, Wang H, Wang X, Liu Q, Ye M, Tan W. A Smart, Photocontrollable Drug Release Nanosystem for Multifunctional Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:5847-5854. [PMID: 28124556 DOI: 10.1021/acsami.6b15414] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Multifunctional synergistic therapy holds promise in biomedical studies and clinical practice. However, strategies aimed at easily integrating the components of such multimodal therapies are needed. Therefore, we herein report a smart drug release nanosystem able to perform photodynamic therapy, photothermal therapy and chemotherapy in a photocontrollable manner. Doxorubicin (DOX), a chemotherapy drug, and 5, 10, 15, 20-tetrakis (1-methylpyridinium-4-yl) porphyrin (TMPyP4), a photosensitizer, were physically intercalated into a DNA assembly immobilized on gold nanorods. The drugs were efficiently delivered to target cells and released under light irradiation, resulting in a synergism that combined phototherapy and chemotherapy for cancer treatment. This smart, photocontrollable drug release nanosystem promises precisely controlled drug release for multifunctional synergistic cancer therapy.
Collapse
Affiliation(s)
- Yi Yi
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
| | - Huijing Wang
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
| | - Xuewei Wang
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
| | - Qiaoling Liu
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, College of Biology, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University , Changsha 410082, China
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Shands Cancer Center, University of Florida Genetics Institute, and McKnight Brain Institute, University of Florida , Gainesville, Florida 32611-7200, United States
| |
Collapse
|