1
|
Dey S, Dinakar YH, R S, Jain V, Jain R. Navigating the therapeutic landscape for breast cancer: targeting breast cancer stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2387-2406. [PMID: 39441235 DOI: 10.1007/s00210-024-03542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Breast cancer is a common and deadly malignancy that affects women globally, and breast cancer stem cells (BCSCs) play an important role in tumorigenesis, development, metastasis, and recurrence. Traditional therapies often fail to eliminate BCSCs, leading to treatment resistance and relapse. This review explores the therapeutic strategies which are designed to target BCSCs, including inhibition of key signaling pathway and targeting receptor. This paper also explores the approaches to targeting BCSCs including chemotherapy, phytomedicines, and nanotechnology. Nanotechnology has gained a lot of importance in cancer therapy because of its ability to deliver therapeutic agents with more precision and minimal side effects. Various chemotherapeutic drugs, siRNAs, or gene editing tools are delivered efficiently with the use of nanocarriers which target pathways, receptors, and proteins associated with BCSCs. Over the past few years, stimuli-responsive and receptor-targeted nanocarriers have been explored for better therapeutic effects. In recent times, strategies such as chimeric antigen receptor (CAR) T-cell therapy, ablation therapy, and cell-free therapies are explored for targeting these stem cells. This review provides a recent developmental overview of strategies to attack BCSCs from conventional chemotherapeutic agents to nanotechnological platforms such as polymeric, lipidic, and metal-based nanoparticles and advanced technologies like CAR T cell therapies.
Collapse
Affiliation(s)
- Soudeep Dey
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Soundarya R
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| |
Collapse
|
2
|
McDonald ES, Pan TC, Pant DK, Troester MA, Kossenkov AV, Mankoff DA, Mach RH, Chodosh LA. Ternary Complex Components Responsible for Rapid LDL Internalization as Biomarkers for Breast Cancer Associated with Proliferation and Early Recurrence. CANCER RESEARCH COMMUNICATIONS 2025; 5:226-239. [PMID: 39804138 PMCID: PMC11791746 DOI: 10.1158/2767-9764.crc-23-0562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/16/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
SIGNIFICANCE This first large-scale analysis of the putative ternary complex responsible for rapid low-density lipoprotein internalization in breast cancer reveals a link between component expression and recurrence, with prognostic implications for identifying patients needing supplemental posttreatment surveillance and/or additional therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth S. McDonald
- Division of Breast Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tien-Chi Pan
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dhruv K. Pant
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melissa A. Troester
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Andrew V. Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - David A. Mankoff
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H. Mach
- Radiochemistry, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lewis A. Chodosh
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Dykman L, Khlebtsov B, Khlebtsov N. Drug delivery using gold nanoparticles. Adv Drug Deliv Rev 2025; 216:115481. [PMID: 39617254 DOI: 10.1016/j.addr.2024.115481] [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: 07/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Modern nanotechnologies provide various possibilities for efficiently delivering drugs to biological targets. This review focuses on using functionalized gold nanoparticles (GNPs) as a drug delivery platform. Owing to their exceptional size and surface characteristics, GNPs are a perfect drug delivery vehicle for targeted and selective distribution. Several in vitro and in vivo tests have shown how simple it is to tailor these particles to administer chemical medications straight to tumors. The GNP surface can also be coated with ligands to modify drug release or improve selectivity. Moreover, the pharmacological activity can be enhanced by using the photothermal characteristics of the particles.
Collapse
Affiliation(s)
- Lev Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Boris Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - Nikolai Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, "Saratov Scientific Centre of the Russian Academy of Sciences", 13 Prospekt Entuziastov, Saratov 410049, Russia; Saratov State University, 83 Ulitsa Astrakhanskaya, Saratov 410012, Russia.
| |
Collapse
|
4
|
Dong Z, Xue K, Verma A, Shi J, Wei Z, Xia X, Wang K, Zhang X. Photothermal therapy: a novel potential treatment for prostate cancer. Biomater Sci 2024; 12:2480-2503. [PMID: 38592730 DOI: 10.1039/d4bm00057a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related death in men, and most PCa patients treated with androgen deprivation therapy will progress to metastatic castration-resistant prostate cancer (mCRPC) due to the lack of efficient treatment. Recently, lots of research indicated that photothermal therapy (PTT) was a promising alternative that provided an accurate and efficient prostate cancer therapy. A photothermic agent (PTA) is a basic component of PPT and is divided into organic and inorganic PTAs. Besides, the combination of PTT and other therapies, such as photodynamic therapy (PDT), immunotherapy (IT), chemotherapy (CT), etc., provides an more efficient strategy for PCa therapy. Here, we introduce basic information about PTT and summarize the PTT treatment strategies for prostate cancer. Based on recent works, we think the combination of PPT and other therapies provides a novel possibility for PCa, especially CRPC clinical treatment.
Collapse
Affiliation(s)
- Zirui Dong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anushikha Verma
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Ave, Wuhan 430022, Hubei, China.
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
5
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
6
|
Li J, Zhang J, Gao Y, Lei S, Wu J, Chen X, Wang K, Duan X, Men K. Targeted siRNA Delivery by Bioinspired Cancer Cell Membrane-Coated Nanoparticles with Enhanced Anti-Cancer Immunity. Int J Nanomedicine 2023; 18:5961-5982. [PMID: 37901359 PMCID: PMC10612485 DOI: 10.2147/ijn.s429036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/07/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Cell-membrane nanocarriers are usually constructed by modifying the nanoparticle surface with cell membrane extracts, which has a direct benefit in endowing targeting capacity to nanocarriers based on their original cell types. However, delivering nucleic acid cargos by cell membrane-based nanoparticles is difficult owing to the strong negative charge of the cell membrane fraction. In this study, we developed a cancer cell membrane-based drug delivery system, the cMDS, for efficient siRNA delivery. Meanwhile, the cancer-specific immune response stimulated by the gene vector itself could offer synergistic anti-cancer ability. Methods The cMDS was prepared by ultrasound, and its transfection efficiency and anti-cancer ability were examined using cultures of CT26 cells. MTT and red blood cell hemolysis tests were performed to assess the safety of cMDS, while its targeted gene delivery and strong immune stimulation were investigated in a subcutaneous tumor model. Moreover, the detailed anti-cancer immune stimulation mechanisms of cMDS are uncovered by protein chip analysis. Results The cMDS was spherical core-shell structure. It showed high transfection efficiency and anti-cancer ability in vitro. In animal experiments, intravenously administered cMDS/siStat3 complex efficiently suppress the growth of colon cancer. Moreover, the result of protein chip analysis suggested that cMDS affect the migration and chemotaxis of immune cells. Conclusion The cMDS shows obvious tumor tissue-specific accumulation properties and strong immune stimulation ability. It is an advanced targeted gene delivery system with potent immunotherapeutic properties.
Collapse
Affiliation(s)
- Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Yan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jieping Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xiaohua Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Kaiyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
7
|
Recent applications of phase-change materials in tumor therapy and theranostics. BIOMATERIALS ADVANCES 2023; 147:213309. [PMID: 36739784 DOI: 10.1016/j.bioadv.2023.213309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Phase-change materials (PCMs) are a type of special material which can store and release a large amount of thermal energy without any significant temperature change. They are emerging in recent years as a promising functional material in tumor therapy and theranostics due to their accurate responses to the temperature variations, biocompatibility and low toxicity. In this review, we will introduce the main types of PCMs and their desirable physiochemical properties for biomedical applications, and highlight the recent progress of PCM's applications in the modulated release of antitumor drugs, with special attentions paid to various ways to initiate temperature-dependent phase change, the concomitant thermal therapy and its combination with or activation of other therapies, particularly unconventional therapies. We will also summarize PCM's recent applications in tumor theranostics, where both drugs and imaging probes are delivered by PCMs for controlled drug release and imaging-guided therapy. Finally, the future perspectives and potential limitations of harnessing PCMs in tumor therapy will be discussed.
Collapse
|
8
|
Cheng H, Wang C, Qin D, Xia Y. Galvanic Replacement Synthesis of Metal Nanostructures: Bridging the Gap between Chemical and Electrochemical Approaches. Acc Chem Res 2023; 56:900-909. [PMID: 36966410 PMCID: PMC10077583 DOI: 10.1021/acs.accounts.3c00067] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
ConspectusGalvanic replacement synthesis involves oxidation and dissolution of atoms from a substrate while the salt precursor to another material with a higher reduction potential is reduced and deposited on the substrate. The driving force or spontaneity of such a synthesis comes from the difference in reduction potential between the redox pairs involved. Both bulk and micro/nanostructured materials have been explored as substrates for galvanic replacement synthesis. The use of micro/nanostructured materials can significantly increase the surface area, offering immediate advantages over the conventional electrosynthesis. The micro/nanostructured materials can also be intimately mixed with the salt precursor in a solution phase, resembling the setting of a typical chemical synthesis. The reduced material tends to be directly deposited on the surface of the substrate, just like the situation in an electrosynthesis. Different from an electrosynthesis where the two electrodes are spatially separated by an electrolyte solution, the cathodes and anodes are situated on the same surface, albeit at different sites, even for a micro/nanostructured substrate. Since the oxidation and dissolution reactions occur at sites different from those for reduction and deposition reactions, one can control the growth pattern of the newly deposited atoms on the same surface of a substrate to access nanostructured materials with diverse and controllable compositions, shapes, and morphologies in a single step. Galvanic replacement synthesis has been successfully applied to different types of substrates, including those made of crystalline and amorphous materials, as well as metallic and nonmetallic materials. Depending on the substrate involved, the deposited material can take different nucleation and growth patterns, resulting in diverse but well-controlled nanomaterials sought for a variety of studies and applications.In this Account, we recapitulate our efforts over the past two decades in fabricating metal nanostructures for a broad range of applications by leveraging the unique capability of galvanic replacement synthesis. We begin with a brief introduction to the fundamentals of galvanic replacement between metal nanocrystals and salt precursors, followed by a discussion of the roles played by surface capping agents in achieving site-selected carving and deposition for the fabrication of various bimetallic nanostructures. Two examples based on the Ag-Au and Pd-Pt systems are selected to illustrate the concept and mechanism. We then highlight our recent work on the galvanic replacement synthesis involving nonmetallic substrates, with a focus on the protocol, mechanistic understanding, and experimental control for the fabrication of Au- and Pt-based nanostructures with tunable morphologies. Finally, we showcase the unique properties and applications of nanostructured materials derived from galvanic replacement reactions for biomedicine and catalysis. We also offer some perspectives on the challenges and opportunities in this emerging field of research.
Collapse
Affiliation(s)
- Haoyan Cheng
- School of Materials Science and Engineering, Henan University of Science and Technology, Luoyang 471023, P. R. China
| | - Chenxiao Wang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Dong Qin
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Younan Xia
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
9
|
Wu Z, Wang J, Zhao L, Li C, Lu Y. A novel donor-acceptor structured diketopyrrolopyrrole-based conjugated polymer synthesized by direct arylation polycondensation (DArP) for highly efficient antimicrobial photothermal therapy. Biomater Sci 2023; 11:2151-2157. [PMID: 36729407 DOI: 10.1039/d2bm02024f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A novel donor (D)-acceptor (A) structured conjugated polymer (PDPP-TP), which contains two alternating D-A pairs, namely thiophene (T)-diketopyrrolopyrrole (DPP) and thiophenen (T)-thieno[3,4-b]pyrazine (TP) along the main chain of the polymer, was synthesized by direct arylation polycondensation (DArP) for a highly efficient photothermal antibacterial treatment. The hydrophilic PDPP-TP-based nanoparticles (PTNPs) with a hydration diameter of about 120 nm were obtained by self-assembly using DSPE-mPEG2000 as the polymer matrix. PTNPs show strong near-infrared (NIR) absorbance with a λmax at 910 nm (ε = 2.25 × 104 L mol-1 cm-1) and NIR light-triggered photoactivity with a high photothermal conversion efficiency (PTCE) of 52.8% under 880 nm laser irradiation. Keeping the merits of excellent biocompatibility and photostability, PTNPs exhibited remarkable bacterial inhibition efficiency of almost 100% against Gram-negative E. coli and Gram-positive S. aureus with the help of an 880 nm laser (0.7 W cm-2, 6 min), demonstrating its great potential as photothermal materials with a broad spectrum of activity for the effective treatment of microbial infections.
Collapse
Affiliation(s)
- Zhihui Wu
- School of Materials Science & Engineering, Tianjin Key Laboratory for Photoelectric Materials and Devices, Key Laboratory of Display Materials &Photoelectric Devices, Ministry of Education, Tianjin University of Technology, Tianjin 300384, China.
| | - Jing Wang
- School of Materials Science & Engineering, Tianjin Key Laboratory for Photoelectric Materials and Devices, Key Laboratory of Display Materials &Photoelectric Devices, Ministry of Education, Tianjin University of Technology, Tianjin 300384, China.
| | - Linlin Zhao
- School of Materials Science & Engineering, Tianjin Key Laboratory for Photoelectric Materials and Devices, Key Laboratory of Display Materials &Photoelectric Devices, Ministry of Education, Tianjin University of Technology, Tianjin 300384, China.
| | - Chenxi Li
- Key Laboratory of Functional Polymer Materials, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yan Lu
- School of Materials Science & Engineering, Tianjin Key Laboratory for Photoelectric Materials and Devices, Key Laboratory of Display Materials &Photoelectric Devices, Ministry of Education, Tianjin University of Technology, Tianjin 300384, China.
| |
Collapse
|
10
|
Liu S, Zhang T, Li S, Wu Q, Wang K, Xu X, Lu M, Shao R, Zhao W, Liu H. Biomimetic Nanobomb for Synergistic Therapy with Inhibition of Cancer Stem Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206503. [PMID: 36587973 DOI: 10.1002/smll.202206503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Cancer stem cells (CSCs), a type of cell with self-renewal, unlimited proliferation, and insensitivity to common physical and chemical factors, are the key to cancer metastasis, recurrence, and chemo-resistance. Available CSCs inhibition strategies are mainly based on small molecule drugs, yet are limited by their off-target toxicity. The link between CSCs and non-CSCs interconversion is difficult to sever. In this work, a nanotherapeutic strategy based on MnOx -loaded polydopamine (MnOx /PDA) nanobombs with chemodynamic, photodynamic, photothermal and biodegradation properties to inhibit CSCs and non-CSCs concurrently is reported. The MnOx /PDA nanobombs can directly disrupt the microenvironment and tumorigenic capacity of CSCs by generating hyperthermia, oxidative stress and alleviating hypoxia. The markers of CSCs are subsequently downregulated, leading to the clearance of CSCs. Meanwhile, the synergistic therapy mediated by MnOx /PDA nanobombs can directly ablate the bulk tumor cells, thus cutting off the supply of CSCs transformation. For tumor targeting, MnOx /PDA is coated with macrophage membrane. The final tumor inhibition rate of the synergistic therapy is 70.8% in colorectal cancer (CRC) model. Taken together, the present work may open up the exploration of nanomaterial-based synergistic therapy for the simultaneous elimination of therapeutically resistant CSCs and non-CSCs.
Collapse
Affiliation(s)
- Shuang Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Tianshu Zhang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shanshan Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kexin Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xican Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - MingZhu Lu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wuli Zhao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
11
|
Lee SS, Paliouras M, Trifiro MA. Functionalized Carbon Nanoparticles as Theranostic Agents and Their Future Clinical Utility in Oncology. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010108. [PMID: 36671680 PMCID: PMC9854994 DOI: 10.3390/bioengineering10010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Over the years, research of nanoparticle applications in pre-clinical and clinical applications has greatly advanced our therapeutic and imaging approaches to many diseases, most notably neoplastic disorders. In particular, the innate properties of inorganic nanomaterials, such as gold and iron oxide, as well as carbon-based nanoparticles, have provided the greatest opportunities in cancer theranostics. Carbon nanoparticles can be used as carriers of biological agents to enhance the therapeutic index at a tumor site. Alternatively, they can also be combined with external stimuli, such as light, to induce irreversible physical damaging effects on cells. In this review, the recent advances in carbon nanoparticles and their use in cancer theranostics will be discussed. In addition, the set of evaluations that will be required during their transition from laboratory investigations toward clinical trials will be addressed.
Collapse
Affiliation(s)
- Seung S. Lee
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute for Medical Research—Jewish General Hospital, Montreal, QC H4A 3J1, Canada
| | - Miltiadis Paliouras
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute for Medical Research—Jewish General Hospital, Montreal, QC H4A 3J1, Canada
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Department of Oncology, McGill University, Montreal, QC H4A 3J1, Canada
- Correspondence:
| | - Mark A. Trifiro
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute for Medical Research—Jewish General Hospital, Montreal, QC H4A 3J1, Canada
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
12
|
Khakbaz F, Mirzaei M, Mahani M. Lecithin sensitized thermo-sensitive niosome using NIR-carbon dots for breast cancer combined chemo-photothermal therapy. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
Popescu MT, Tsitsilianis C. Gold/Pentablock Terpolymer Hybrid Multifunctional Nanocarriers for Controlled Delivery of Tamoxifen: Effect of Nanostructure on Release Kinetics. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123764. [PMID: 35744890 PMCID: PMC9231331 DOI: 10.3390/molecules27123764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022]
Abstract
Here, we describe the preparation and characterization of organic/inorganic hybrid polymer multifunctional nanocarriers. Novel nanocomposites of gold nanoparticles using pH-responsive coordination pentablock terpolymers of poly(ε-caprolactone)-b-poly(ethylene oxide)-b-poly(2-vinylpyridine)-b-poly(ethylene oxide)-b-poly(ε-caprolactone), bearing or not bearing partially quaternized vinylpyridine moieties, were studied. The template morphology of the coordination pentablock terpolymer at physiological pH ranges from crew-cut to multicompartmentalized micelles which can be tuned by chemical modification of the central block. Additionally, the presence of 2VP groups allows the coordination of gold ions, which can be reduced in situ to construct gold@polymer nanohybrids. Furthermore, the possibility of tuning the gold distribution in the micelles, through partial quaternization of the central P2VP block, was also investigated. Various morphological gold colloidal nanoparticles such as gold@core-corona nanoparticles and gold@core-gold@corona nanoparticles were synthesized on the corresponding template of the pentablock terpolymer, first by coordination with gold ions, followed by reduction with NaBH4. The pentablock and gold@pentablock nanoparticles could sparingly accommodate a water-soluble drug, Tamoxifen (TAX), in their hydrophobic micellar cores. The nanostructure of the nanocarrier remarkably affects the TAX delivery kinetics. Importantly, the hybrid gold@polymer nanoparticles showed prolonged release profiles for the guest molecule, relative to the corresponding bare amphiphilic pentablock polymeric micelles. These Gold@pentablock terpolymer hybrid nanoparticles could act as a multifunctional theranostic nanoplatform, integrating sustainable pH-controlled drug delivery, diagnostic function and photothermal therapy.
Collapse
|
14
|
Pan Y, Zhou S, Liu C, Ma X, Xing J, Parshad B, Li W, Wu A, Haag R. Dendritic Polyglycerol-Conjugated Gold Nanostars for Metabolism Inhibition and Targeted Photothermal Therapy in Breast Cancer Stem Cells. Adv Healthc Mater 2022; 11:e2102272. [PMID: 34990518 PMCID: PMC11468648 DOI: 10.1002/adhm.202102272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer stem cells (CSCs) are believed to be responsible for tumor initiation, invasion, metastasis, and recurrence, which lead to treatment failure. Thus, developing effective CSC-targeted therapeutic strategies is crucial for enhancing therapeutic efficacy. In this work, GNSs-dPG-3BP, TPP, and HA nanocomposite particles are developed by simultaneously conjugating hexokinase 2 (HK2) inhibitor 3-bromopyruvate (3BP), mitochondrial targeting molecule triphenyl phosphonium (TPP), and CSCs targeting agent hyaluronic acid (HA) onto gold nanostars-dendritic polyglycerol (GNSs-dPG) nanoplatforms for efficient eradication of CSCs. The nanocomposite particles possess good biocompatibility and exhibit superior mitochondrial-bound HK2 binding ability via 3BP to inhibit metabolism, and further induce cellular apoptosis by releasing the cytochrome c. Therefore, it enhanced the therapeutic efficacy of CSCs-specific targeted photothermal therapy (PTT), and achieved a synergistic effect for the eradication of breast CSCs. After administration of the synergistic treatment, the self-renewal of breast CSCs and the stemness gene expression are suppressed, CSC-driven mammosphere formation is diminished, the in vivo tumor growth is effectively inhibited, and CSCs are eradicated. Altogether, GNSs-dPG-3BP, TPP, and HA nanocomposite particles have been developed, which will provide a novel strategy for precise and highly efficient targeted eradication of CSCs.
Collapse
Affiliation(s)
- Yuanwei Pan
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Suqiong Zhou
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Chuang Liu
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Xuehua Ma
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Jie Xing
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Badri Parshad
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Wenzhong Li
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Aiguo Wu
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| |
Collapse
|
15
|
Makvandi P, Chen M, Sartorius R, Zarrabi A, Ashrafizadeh M, Dabbagh Moghaddam F, Ma J, Mattoli V, Tay FR. Endocytosis of abiotic nanomaterials and nanobiovectors: Inhibition of membrane trafficking. NANO TODAY 2021; 40:101279. [PMID: 34518771 PMCID: PMC8425779 DOI: 10.1016/j.nantod.2021.101279] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Humans are exposed to nanoscopical nanobiovectors (e.g. coronavirus SARS-CoV-2) as well as abiotic metal/carbon-based nanomaterials that enter cells serendipitously or intentionally. Understanding the interactions of cell membranes with these abiotic and biotic nanostructures will facilitate scientists to design better functional nanomaterials for biomedical applications. Such knowledge will also provide important clues for the control of viral infections and the treatment of virus-induced infectious diseases. In the present review, the mechanisms of endocytosis are reviewed in the context of how nanomaterials are uptaken into cells. This is followed by a detailed discussion of the attributes of man-made nanomaterials (e.g. size, shape, surface functional groups and elasticity) that affect endocytosis, as well as the different human cell types that participate in the endocytosis of nanomaterials. Readers are then introduced to the concept of viruses as nature-derived nanoparticles. The mechanisms in which different classes of viruses interact with various cell types to gain entry into the human body are reviewed with examples published over the last five years. These basic tenets will enable the avid reader to design advanced drug delivery and gene transfer nanoplatforms that harness the knowledge acquired from endocytosis to improve their biomedical efficacy. The review winds up with a discussion on the hurdles to be addressed in mimicking the natural mechanisms of endocytosis in nanomaterials design.
Collapse
Affiliation(s)
- Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Meiling Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Naples 80131, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Farnaz Dabbagh Moghaddam
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Jingzhi Ma
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Virgilio Mattoli
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
16
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
17
|
Qiu J, Liu Y, Xia Y. Radiolabeling of Gold Nanocages for Potential Applications in Tracking, Diagnosis, and Image-Guided Therapy. Adv Healthc Mater 2021; 10:e2002031. [PMID: 33470560 PMCID: PMC8289932 DOI: 10.1002/adhm.202002031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/06/2021] [Indexed: 12/23/2022]
Abstract
Gold nanocages (AuNCs) have emerged as a novel class of multifunctional nanomaterials with an array of applications in nanomedicine, including drug delivery, controlled release, as well as disease diagnosis and treatment. Labeling AuNCs with radionuclides not only offers additional therapeutic capabilities but also makes it easy to analyze their biodistribution, monitor their uptake by the tissue or organ of interest, and optimize their performance in both diagnosis and treatment. Here, an introduction to the chemical synthesis and optical properties of AuNCs is provided in the beginning. The methods developed for their radiolabeling are then showcased, followed by the use of radiolabeled AuNCs in tracking and quantifying their pharmacokinetics, including biodistribution, tumor uptake, and intratumoral distribution. Finally, their potential applications in targeted imaging and image-guided therapy are discussed.
Collapse
Affiliation(s)
- Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
18
|
Qiu J, Xu J, Xia Y. Nanobottles for Controlled Release and Drug Delivery. Adv Healthc Mater 2021; 10:e2000587. [PMID: 32543127 PMCID: PMC7738374 DOI: 10.1002/adhm.202000587] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/06/2020] [Indexed: 12/22/2022]
Abstract
Nanobottles refer to colloidal particles with a hollow interior and a single opening in the wall. These unique features make them ideal carriers for the loading, encapsulation, release, and delivery of various types of theranostic agents in an array of biomedical applications. The hollow interior gives them a high loading capacity while the opening enables quick loading and controlled release of the payload(s). More significantly, on-demand release can be readily achieved by adding a stimuli-responsive material as the inner matrix or cork stopper. This progress report begins with an introduction to the general structures and properties of nanobottles, followed by a brief discussion on the methods developed for their fabrication. The use of nanobottles for loading different types of payloads is then showcased, including small-molecule drugs, biomacromolecules, imaging contrast agents, and functional nanoparticles. The strategies explored for controlling the release by varying the size of the opening and/or integrating with a stimuli-responsive material are also highlighted. This paper concludes with some perspectives on future directions for this class of nanomaterials in terms of fabrication, functionalization, and application.
Collapse
Affiliation(s)
- Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Jianchang Xu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
19
|
Duan H, Liu Y, Gao Z, Huang W. Recent advances in drug delivery systems for targeting cancer stem cells. Acta Pharm Sin B 2021; 11:55-70. [PMID: 33532180 PMCID: PMC7838023 DOI: 10.1016/j.apsb.2020.09.016] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/25/2020] [Accepted: 07/12/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a subpopulation of cancer cells with functions similar to those of normal stem cells. Although few in number, they are capable of self-renewal, unlimited proliferation, and multi-directional differentiation potential. In addition, CSCs have the ability to escape immune surveillance. Thus, they play an important role in the occurrence and development of tumors, and they are closely related to tumor invasion, metastasis, drug resistance, and recurrence after treatment. Therefore, specific targeting of CSCs may improve the efficiency of cancer therapy. A series of corresponding promising therapeutic strategies based on CSC targeting, such as the targeting of CSC niche, CSC signaling pathways, and CSC mitochondria, are currently under development. Given the rapid progression in this field and nanotechnology, drug delivery systems (DDSs) for CSC targeting are increasingly being developed. In this review, we summarize the advances in CSC-targeted DDSs. Furthermore, we highlight the latest developmental trends through the main line of CSC occurrence and development process; some considerations about the rationale, advantages, and limitations of different DDSs for CSC-targeted therapies were discussed.
Collapse
Key Words
- ABC, ATP binding cassette
- AFN, apoferritin
- ALDH, aldehyde dehydrogenase
- BM-MSCs-derived Exos, bone marrow mesenchymal stem cells-derived exosomes
- Biomarker
- CAFs, cancer-associated fibroblasts
- CL-siSOX2, cationic lipoplex of SOX2 small interfering RNA
- CMP, carbonate-mannose modified PEI
- CQ, chloroquine
- CSCs, cancer stem cells
- Cancer stem cells
- Cancer treatment
- Cellular level
- DCLK1, doublecortin-like kinase 1
- DDSs, drug delivery systems
- DLE, drug loading efficiency
- DOX, doxorubicin
- DQA-PEG2000-DSPE, dequlinium and carboxyl polyethylene glycol-distearoylphosphatidylethanolamine
- Dex, dexamethasone
- Drug delivery systems
- ECM, extracellular matrix
- EMT, epithelial–mesenchymal transition
- EPND, nanodiamond-Epirubicin drug complex
- EpCAM, epithelial cell adhesion molecule
- GEMP, gemcitabine monophosphate
- GLUT1, glucose ligand to the glucose transporter 1
- Glu, glucose
- HCC, hepatocellular carcinoma
- HH, Hedgehog
- HIF1α, hypoxia-inducible factor 1-alpha
- HNSCC, head and neck squamous cell carcinoma
- IONP, iron oxide nanoparticle
- LAC, lung adenocarcinoma
- LNCs, lipid nanocapsules
- MAPK, mitogen-activated protein kinase
- MB, methylene blue
- MDR, multidrug resistance
- MNP, micellar nanoparticle
- MSNs, mesoporous silica nanoparticles
- Molecular level
- NF-κB, nuclear factor-kappa B
- Nav, navitoclax
- Niche
- PBAEs, poly(β-aminoester)
- PDT, photodynamic therapy
- PEG-PCD, poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate-graft-dodecanol)
- PEG-PLA, poly(ethylene glycol)-b-poly(d,l-lactide)
- PEG-b-PLA, poly(ethylene glycol)-block-poly(d,l-lactide)
- PLGA, poly(ethylene glycol)-poly(d,l-lactide-co-glycolide)
- PTX, paclitaxel
- PU-PEI, polyurethane-short branch-polyethylenimine
- SLNs, solid lipid nanoparticles
- SSCs, somatic stem cells
- Sali-ABA, 4-(aminomethyl) benzaldehyde-modified Sali
- TNBC, triple negative breast cancer
- TPZ, tirapazamine
- Targeting strategies
- cRGD, cyclic Arg-Gly-Asp
- iTEP, immune-tolerant, elastin-like polypeptide
- mAbs, monoclonal antibodies
- mPEG-b-PCC-g-GEM-g-DC-g-CAT, poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylenecarbonate-graft-dodecanol-graft-cationic ligands)
- ncRNA, non-coding RNAs
- uPAR, urokinase plasminogen activator receptor
Collapse
Affiliation(s)
- Hongxia Duan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Li S, Shi X, Wang H, Xiao L. A multifunctional dual-shell magnetic nanocomposite with near-infrared light response for synergistic chemo-thermal tumor therapy. J Biomed Mater Res B Appl Biomater 2020; 109:841-852. [PMID: 33135302 DOI: 10.1002/jbm.b.34749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/26/2020] [Accepted: 10/06/2020] [Indexed: 01/29/2023]
Abstract
The synergistic tumor therapy in single nanoplatform has always been the goal for high efficacy tumor treatment while still remains great challenge. This paper reports a versatile nanotheranostic platform enlisting magnetic iron oxide nanoparticles, polydopamine (PDA), gold nanocages (Au nanocage) and metal organic framework (MOF, MIL101-NH2 ) in order to achieve synergistic chemothermal tumor therapy both in vitro and in vivo. The prepared magnetic photothermal nanoparticles (MPNPs) exhibit high drug loading capacity (31.34 mg/g), superior photo-thermal capacity (11.5°C enhancement in 180 s), low bio-toxicity, good magnetic resonance with a low dosage of 22 μg/g, as well as high antitumor efficacy in vivo. Such a novel and multifunctional nanoplatform is expected to find promising applications in target tumor synergistic therapy.
Collapse
Affiliation(s)
- Sheng Li
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China.,School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, PR China
| | - Xiaowen Shi
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China
| | - Haibo Wang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan, PR China
| | - Ling Xiao
- School of Resource and Environmental Science, Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, PR China
| |
Collapse
|
21
|
Qiu J, Huo D, Xia Y. Phase-Change Materials for Controlled Release and Related Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000660. [PMID: 32383215 PMCID: PMC7473464 DOI: 10.1002/adma.202000660] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 05/07/2023]
Abstract
Phase-change materials (PCMs) have emerged as a novel class of thermo-responsive materials for controlled release, where the payloads encapsulated in a solid matrix are released only upon melting the PCM to trigger a solid-to-liquid phase transition. Herein, the advances over the past 10 years in utilizing PCMs as a versatile platform for the encapsulation and release of various types of therapeutic agents and biological effectors are highlighted. A brief introduction to PCMs in the context of desired properties for controlled release and related applications is provided. Among the various types of PCMs, a specific focus is placed on fatty acids and fatty alcohols for their natural availability, low toxicity, biodegradability, diversity, high abundance, and low cost. Then, various methods capable of processing PCMs, and their mixtures with payloads, into stable suspensions of colloidal particles, and the different means for triggering the solid-to-liquid phase transition are discussed. Finally, a range of applications enabled by the controlled release system based on PCMs are presented together with some perspectives on future directions.
Collapse
Affiliation(s)
- Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Da Huo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
22
|
Qiu J, Xie M, Wu T, Qin D, Xia Y. Gold nanocages for effective photothermal conversion and related applications. Chem Sci 2020. [DOI: 10.1039/d0sc05146b] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Gold nanocages are highly effective in converting light to heat, making them versatile for an array of photothermal applications.
Collapse
Affiliation(s)
- Jichuan Qiu
- The Wallace H. Coulter Department of Biomedical Engineering
- Georgia Institute of Technology and Emory University
- Atlanta
- USA
| | - Minghao Xie
- School of Chemistry and Biochemistry
- Georgia Institute of Technology
- Atlanta
- USA
| | - Tong Wu
- The Wallace H. Coulter Department of Biomedical Engineering
- Georgia Institute of Technology and Emory University
- Atlanta
- USA
| | - Dong Qin
- School of Materials Science and Engineering
- Georgia Institute of Technology
- Atlanta
- USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering
- Georgia Institute of Technology and Emory University
- Atlanta
- USA
- School of Chemistry and Biochemistry
| |
Collapse
|
23
|
Yu J, Wang X, Feng J, Meng X, Bu X, Li Y, Zhang N, Wang P. Antimonene Nanoflakes: Extraordinary Photoacoustic Performance for High-Contrast Imaging of Small Volume Tumors. Adv Healthc Mater 2019; 8:e1900378. [PMID: 31290279 DOI: 10.1002/adhm.201900378] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/24/2019] [Indexed: 12/31/2022]
Abstract
Photoacoustic imaging (PAI) has evolved to a stage that high-performance exogeneous contrast agents are urgently needed for imminent biomedical and clinical applications. Given that a material meets the basic criteria of efficient photoacoustic conversion, high biocompatibility, and fast excretion, great effort has been devoted to evaluating various materials for developing advantageous contrast agents to explore the full potentials of PAI. One focus is through modification of the current agents to boost their PA performance; whilst the other focus is to develop novel agents. Antimonene (AM) has emerged as a promising candidate for next generation of electronics among 2D materials due to its outstanding properties. Herein, it is reported that liquid-phase exfoliated antimonene exhibits extraordinary photoacoustic performance, which is not only more advantageous than other 2D materials, such as black phosphorus, graphene oxide, and transition metal dichalcogenides, but also superior to the commonly used PA contrast agents, such as ICG and gold nanorods. An insight analysis reveals that the unique thermal property of AM, including intrinsic low thermal conductivity and the morphology-related high interfacial thermal conductivity, might interpret the high photothermal conversion efficiency, and thus the excellent photoacoustic performance. The prodigious performance allows sensitive monitoring of intracellular events and high-quality in vivo tumor imaging.
Collapse
Affiliation(s)
- Jingwen Yu
- Laboratory for Biomedical PhotonicsBeijing University of Technology Beijing 100124 China
- Laboratory for Advanced Laser Technology and ApplicationsInstitute of Laser EngineeringBeijing University of Technology Beijing 100124 China
- Key Laboratory of Trans‐Scale Laser Manufacturing TechnologyMinistry of Education China Beijing 100124 China
- Beijing Engineering Research Center of Laser Technology Beijing 100124 China
| | - Xiu‐Hong Wang
- Laboratory for Biomedical PhotonicsBeijing University of Technology Beijing 100124 China
- Laboratory for Advanced Laser Technology and ApplicationsInstitute of Laser EngineeringBeijing University of Technology Beijing 100124 China
- Key Laboratory of Trans‐Scale Laser Manufacturing TechnologyMinistry of Education China Beijing 100124 China
- Beijing Engineering Research Center of Laser Technology Beijing 100124 China
| | - Jinchao Feng
- College of Electronic Information and Control EngineeringBeijing University of Technology Beijing 100124 China
| | - Xiaotong Meng
- Department of BioengineeringImperial College London London SW7 2BP UK
| | - Xiangbao Bu
- Laboratory for Advanced Laser Technology and ApplicationsInstitute of Laser EngineeringBeijing University of Technology Beijing 100124 China
- Key Laboratory of Trans‐Scale Laser Manufacturing TechnologyMinistry of Education China Beijing 100124 China
- Beijing Engineering Research Center of Laser Technology Beijing 100124 China
| | - Yang Li
- Laboratory for Biomedical PhotonicsBeijing University of Technology Beijing 100124 China
- Laboratory for Advanced Laser Technology and ApplicationsInstitute of Laser EngineeringBeijing University of Technology Beijing 100124 China
- Key Laboratory of Trans‐Scale Laser Manufacturing TechnologyMinistry of Education China Beijing 100124 China
- Beijing Engineering Research Center of Laser Technology Beijing 100124 China
| | - Na Zhang
- College of Electronic Information and Control EngineeringBeijing University of Technology Beijing 100124 China
| | - Pu Wang
- Laboratory for Advanced Laser Technology and ApplicationsInstitute of Laser EngineeringBeijing University of Technology Beijing 100124 China
- Key Laboratory of Trans‐Scale Laser Manufacturing TechnologyMinistry of Education China Beijing 100124 China
- Beijing Engineering Research Center of Laser Technology Beijing 100124 China
| |
Collapse
|
24
|
Sharifi M, Attar F, Saboury AA, Akhtari K, Hooshmand N, Hasan A, El-Sayed MA, Falahati M. Plasmonic gold nanoparticles: Optical manipulation, imaging, drug delivery and therapy. J Control Release 2019; 311-312:170-189. [PMID: 31472191 DOI: 10.1016/j.jconrel.2019.08.032] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/25/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022]
Abstract
Over the past two decades, the development of plasmonic nanoparticle (NPs), especially gold (Au) NPs, is being pursued more seriously in the medical fields such as imaging, drug delivery, and theranostic systems. However, there is no comprehensive review on the effect of the physical and chemical parameters of AuNPs on their plasmonic properties as well as the use of these unique characteristic in medical activities such as imaging and therapeutics. Therefore, in this literature the surface plasmon resonance (SPR) modeling of AuNPs was accurately captured toward precision medicine. Indeed, we investigated the importance of plasmonic properties of AuNPs in optical manipulation, imaging, drug delivery, and photothermal therapy (PTT) of cancerous cells based on their physicochemical properties. Finally, some challenges regarding the commercialization of AuNPs in future medicine such as, cytotoxicity, lack of standards for medical applications, high cost, and time-consuming process were discussed.
Collapse
Affiliation(s)
- Majid Sharifi
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Farnoosh Attar
- Department of Biology, Faculty of Food Industry & Agriculture, Standard Research Institute, Karaj, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Nasrin Hooshmand
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Mostafa A El-Sayed
- Laser Dynamics Laboratory, School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
25
|
Qiu W, Zhang H, Chen X, Song L, Cui W, Ren S, Wang Y, Guo K, Li D, Chen R, Wang Z. A GPC1-targeted and gemcitabine-loaded biocompatible nanoplatform for pancreatic cancer multimodal imaging and therapy. Nanomedicine (Lond) 2019; 14:2339-2353. [PMID: 31414945 DOI: 10.2217/nnm-2019-0063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: Biomarker-targeted nanocarrier holds promise for early diagnosis and effective therapy of cancer. Materials & methods: This work successfully designs and evaluates GPC1-targeted, gemcitabine (GEM)-loaded multifunctional gold nanocarrier for near-infrared fluorescence (NIRF)/MRI and targeted chemotherapy against pancreatic cancer in vitro and in vivo. Results: Blood biochemical and histological analyses show that the in vivo toxicity of GPC1-GEM-nanoparticles (NPs) was negligible. Both in vitro and in vivo studies demonstrate that GPC1-GEM-NPs can be used as NIRF/MR contrast agent for pancreatic cancer detection. Treatment of xenografted mice with GPC1-GEM-NPs shows a higher tumor inhibitory effect compared with controls. Conclusion: This novel theranostic nanoplatform provides early diagnostic and effective therapeutic potential for pancreatic cancer.
Collapse
Affiliation(s)
- Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Huifeng Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Lina Song
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Wenjing Cui
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Shuai Ren
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Yajie Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Kai Guo
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rong Chen
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, MD 21201, USA
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| |
Collapse
|
26
|
Qin Z, Du T, Zheng Y, Luo P, Zhang J, Xie M, Zhang Y, Du Y, Yin L, Cui D, Lu Q, Lu M, Wang X, Jiang H. Glutathione Induced Transformation of Partially Hollow Gold-Silver Nanocages for Cancer Diagnosis and Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902755. [PMID: 31347262 DOI: 10.1002/smll.201902755] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Gold-silver nanocages (GSNCs) are widely used in cancer imaging and therapy due to excellent biocompatibility, internal hollow structures, and tunable optical properties. However, their possible responses toward the tumor microenvironment are still not well understood. In this study, it is demonstrated that a kind of relatively small sized (35 nm) and partially hollow GSNCs (absorbance centered at 532 nm) can enhance the intrinsic photoacoustic imaging performances for blood vessels around tumor sites. More importantly, the high concentration of glutathione around the tumor cells' microenvironment may induce the aggregation, disintegration, and agglomeration of these GSNCs sequentially, allowing significant shifts in the absorbance spectrum of GSNCs to the near-infrared (NIR) region. This enhanced absorbance in the NIR region entails the significant photothermal therapy (PTT) effect. In vivo experiments, including photoacoustic microscopy (PAM) for cancer diagnosis and PTT in tumor model mice, also show coincident consequences. Taken together, the slightly hollow GSNCs may assist PAM-based tumor diagnosis and induce a tumor targeted PTT effect. This work paves a new avenue for the development of an alternative tumor diagnostic and therapeutic strategy.
Collapse
Affiliation(s)
- Zhaojian Qin
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Tianyu Du
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Youkun Zheng
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Peng Luo
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Jialei Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Mengyang Xie
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Ying Zhang
- School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Ying Du
- School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Lihong Yin
- School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Daxiang Cui
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiaotong University, Shanghai, 200240, P. R. China
| | - Qiangbing Lu
- National Laboratory of Solid State Microstructures & Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, P. R. China
| | - Minghui Lu
- National Laboratory of Solid State Microstructures & Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, P. R. China
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| |
Collapse
|
27
|
Zhan C, Huang Y, Lin G, Huang S, Zeng F, Wu S. A Gold Nanocage/Cluster Hybrid Structure for Whole-Body Multispectral Optoacoustic Tomography Imaging, EGFR Inhibitor Delivery, and Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900309. [PMID: 31245925 DOI: 10.1002/smll.201900309] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/25/2019] [Indexed: 06/09/2023]
Abstract
Gold nanocages (AuNCs) and gold nanoclusters (AuClusters) are two classes of advantageous nanostructures with special optical properties, and many other attractive properties. Integrating them into one nanosystem may achieve greater and smarter performance. Herein, a hybrid gold nanostructure for fluorescent and optoacoustic tomography imaging, controlled release of drugs, and photothermal therapy (PTT) is demonstrated. For this nanodrug (EA-AB), an epidermal growth factor receptor (EGFR) inhibitor erlotinib (EB) is loaded into AuNCs, which are then capped and functionalized by biocompatible AuCluster@BSA (BSA = bovine serum albumin) conjugates via electrostatic interaction. Upon cell internalization, the lysosomal proteases and low pH cause the release of EB from EA-AB, and also induce fluorescence restoration of the AuCluster for imaging. Irradiation with near-infrared light further promotes the drug release and affords a PTT effect as well. The AuNC-based nanodrug is optoacoustically active, and its biodistribution and metabolic process have been successfully monitored by whole-body and 3D multispectral optoacoustic tomography imaging. Owing to the combined actions of PTT and EGFR pathway blockage, EA-AB exhibits marked tumor inhibition efficacy in vivo.
Collapse
Affiliation(s)
- Chenyue Zhan
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Yong Huang
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Guifang Lin
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Shuailing Huang
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Fang Zeng
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Shuizhu Wu
- State Key Laboratory of Luminescent Materials and Devices, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| |
Collapse
|
28
|
Huang W, Zhao H, Wan J, Zhou Y, Xu Q, Zhao Y, Yang X, Gan L. pH- and photothermal-driven multistage delivery nanoplatform for overcoming cancer drug resistance. Theranostics 2019; 9:3825-3839. [PMID: 31281516 PMCID: PMC6587350 DOI: 10.7150/thno.33958] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 05/08/2019] [Indexed: 01/04/2023] Open
Abstract
Reversing multidrug resistance (MDR) remains a big challenge in cancer therapy. Combining the hyperthermia and chemotherapy is a promising strategy for efficient cancer treatment with MDR reversal. Gold nanocages (GNCs) are an ideal photothermal (PTT)-chemotherapy integration platform due to their good photothermal conversion efficiency and the unique hollow interiors. However, insufficient tumor cell internalization and in vivo premature drug leakage restrict the anticancer activity of GNCs-based drug delivery systems. Methods: pH low insertion peptide (pHLIP)- and thermoresponsive poly(di(ethylene glycol) methyl ether methacrylate-co-oligo(ethylene glycol) methyl ether methacrylate) polymer-conjugated GNCs were rationally constructed to load anticancer drug doxorubicin (DOX@pPGNCs). Tumor acidic environment-responsive tumor cell internalization, and near-infrared (NIR) laser-induced tumor accumulation, penetration and on-demand drug release were systematically examined. Results: DOX@pPGNCs display good photothermal efficacy and thermoresponsive property. NIR laser irradiations at the tumor site significantly enhance tumor accumulation and penetration. Once DOX@pPGNCs reach the tumor site, the conformational transformation of pHLIP at the acidic tumor microenvironment contributes to the enhanced cellular internalization. Furthermore, NIR laser-triggered photothermal effects induce the shrinkage of thermoresponsive polymer, resulting in the opening of the pores of GNCs and a rapid intracellular DOX release to the nuclei. DOX@pPGNCs exhibit synergistic antitumor effect with MDR reversal in vitro and in vivo. Conclusion: DOX@pPGNCs present strong potential to overcome MDR in cancer.
Collapse
Affiliation(s)
- Wenjing Huang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hao Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiangshan Wan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yang Zhou
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingbo Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yanbing Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
29
|
Chen Q, Huo D, Cheng H, Lyu Z, Zhu C, Guan B, Xia Y. Near-Infrared-Triggered Release of Ca 2+ Ions for Potential Application in Combination Cancer Therapy. Adv Healthc Mater 2019; 8:e1801113. [PMID: 30393986 DOI: 10.1002/adhm.201801113] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/23/2018] [Indexed: 12/25/2022]
Abstract
Calcium ion (Ca2+ ), an abundant species in the body, is a potential therapeutic ion with manageable side effects. However, the delivery of such a highly charged species represents a great challenge. Here, a nanosystem based on Au nanocages (AuNCs) and a phase-change material (PCM) for delivering calcium chloride (CaCl2 ) into cancer cells and thereby triggering cell death upon near-infrared (NIR) irradiation is demonstrated. In the absence of NIR irradiation, the nanosystem, denoted CaCl2 -PCM-AuNC, shows negligible cytotoxicity because the Ca2+ ions are fully encapsulated in a solid matrix. Upon NIR irradiation, the Ca2+ ions are swiftly released due to the melting of PCM matrix in response to photothermal heating. The sudden increase in intracellular Ca2+ causes disruption to the mitochondrial Ca2+ homeostasis and thus the loss of mitochondrial membrane potential, subsequently resulting in cell apoptosis. This nanosystem provides a new method for cancer treatment by tightly managing the intracellular concentration of a physiologically essential element.
Collapse
Affiliation(s)
- Qiaoshan Chen
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
- Department of Environmental EngineeringZhejiang University Hangzhou Zhejiang 310058 China
| | - Da Huo
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Haoyan Cheng
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Zhiheng Lyu
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
| | - Baohong Guan
- Department of Environmental EngineeringZhejiang University Hangzhou Zhejiang 310058 China
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University Atlanta GA 30332 USA
- School of Chemistry and BiochemistryGeorgia Institute of Technology Atlanta GA 30332 USA
| |
Collapse
|
30
|
Li H, Li H, Yu W, Huang S, Liu Y, Zhang N, Yuan J, Xu X, Duan S, Hu Y. PEGylated hyaluronidase/NIR induced drug controlled release system for synergetic chemo-photothermal therapy of hepatocellular carcinoma. Eur J Pharm Sci 2019; 133:127-136. [PMID: 30779981 DOI: 10.1016/j.ejps.2019.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/26/2019] [Accepted: 02/15/2019] [Indexed: 01/10/2023]
Abstract
In recent years, cancer treatment has been facing the challenge of increasing antitumor efficiency and avoiding severe adverse effects simultaneously. In this study, we designed a controlled release drug delivery system, doxorubicin (Dox)-loaded and hyaluronic acid (HA)-modified PEGylated gold nanocages (AuNCs), which was designated as PEG-HAn-AuNCs/Dox (n represented 10n HA repeating units were modified on each AuNC). In this system, AuNCs were applied as the photothermal cores, Dox was employed as the model drug, HA was applied as the tumor-microenvironment responsive switch to achieve controlled release, and poly (ethylene glycol) (PEG) was used as the stealth polymer to prolong systemic circulation time. Firstly, we evaluated the physical and chemical properties of the PEG-HAn-AuNCs/Dox with different ratios of HA to AuNC and found that PEG-HA4-AuNCs/Dox was the optimal. Secondly, PEG-HA4-AuNCs/Dox revealed the feature of controlled release, namely, the drug release was triggered by hyaluronidase (HAase) and accelerated by the acidic pH and near-infrared (NIR) irradiation. And then PEG-HA4-AuNCs/Dox could be effectively delivered to a cultured SMMC-7721 cell line in vitro and the tumor tissues of the subcutaneous mouse models of hepatocellular carcinoma (HCC) in vivo. Finally, the results demonstrated the synergetic therapy, namely the combination of chemotherapy and photothermal therapy (PTT) (defined as chemo-photothermal therapy) mediated by PEG-HA4-AuNCs/Dox, could efficiently inhibit the tumor growth both in vitro and in vivo. Therefore, the advantages of PEG-HA4-AuNCs/Dox endowed it as a great potential candidate for HCC treatment.
Collapse
Affiliation(s)
- Huili Li
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Huanjie Li
- General Hospital of Xuzhou Mining Group, Quanshan District Coal Road No. 32, Xuzhou, 221006, PR China
| | - Wei Yu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shengnan Huang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ying Liu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ningxia Zhang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Jinxiu Yuan
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Xin Xu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shaofeng Duan
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China; Henan International Joint Laboratory for Nuclear Protein Regulation, School of Medical Sciences, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China.
| | - Yurong Hu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China; Key Laboratory of Key Technology of Drug Preparation, Ministry of Education, Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| |
Collapse
|
31
|
Poudel BK, Soe ZC, Ruttala HB, Gupta B, Ramasamy T, Thapa RK, Gautam M, Ou W, Nguyen HT, Jeong JH, Jin SG, Choi HG, Yong CS, Kim JO. In situ fabrication of mesoporous silica-coated silver-gold hollow nanoshell for remotely controllable chemo-photothermal therapy via phase-change molecule as gatekeepers. Int J Pharm 2018; 548:92-103. [DOI: 10.1016/j.ijpharm.2018.06.056] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 10/28/2022]
|
32
|
Combination cancer treatment through photothermally controlled release of selenous acid from gold nanocages. Biomaterials 2018; 178:517-526. [DOI: 10.1016/j.biomaterials.2018.03.058] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/14/2018] [Accepted: 03/31/2018] [Indexed: 12/25/2022]
|
33
|
Sigma-2 receptor: past, present and perspectives on multiple therapeutic exploitations. Future Med Chem 2018; 10:1997-2018. [DOI: 10.4155/fmc-2018-0072] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Identification of sigma-2 receptor (sig-2R) has been controversial. Nevertheless, interest in sig-2R is high for its overexpression in tumors and potentials in oncology. Additionally, sig-2R antagonists inhibit Aβ binding at neurons, blocking the cognitive impairments of Alzheimer's disease. The most representative classes of sig-2R ligands are herein treated with focus on compounds that served to study sig-2R biology and to produce sig-2R: fluorescent ligands; multifunctional anticancer agents; and targeting nanoparticles. Although fluorescent ligands serve as ‘green’ pharmacological tools, sig-2R-multifunctional conjugates and sig-2R-targeted nanoparticles show how sig-2R targeting increases the activity of anticancer drugs in tumors with reduced toxicity. Altogether, this review draws a picture of the multiple approaches of sig-2R ligands in cancer therapy and as Alzheimer's disease modifying disease agents.
Collapse
|
34
|
Roper DK, Berry KR, Dunklin JR, Chambers C, Bejugam V, Forcherio GT, Lanier M. Effects of geometry and composition of soft polymer films embedded with nanoparticles on rates for optothermal heat dissipation. NANOSCALE 2018; 10:11531-11543. [PMID: 29892737 DOI: 10.1039/c8nr00977e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Embedding soft matter with nanoparticles (NPs) can provide electromagnetic tunability at sub-micron scales for a growing number of applications in healthcare, sustainable energy, and chemical processing. However, the use of NP-embedded soft material in temperature-sensitive applications has been constrained by difficulties in validating the prediction of rates for energy dissipation from thermally insulating to conducting behavior. This work improved the embedment of monodisperse NPs to stably decrease the inter-NP spacings in polydimethylsiloxane (PDMS) to nano-scale distances. Lumped-parameter and finite element analyses were refined to apportion the effects of the structure and composition of the NP-embedded soft polymer on the rates for conductive, convective, and radiative heat dissipation. These advances allowed for the rational selection of PDMS size and NP composition to optimize measured rates of internal (conductive) and external (convective and radiative) heat dissipation. Stably reducing the distance between monodisperse NPs to nano-scale intervals increased the overall heat dissipation rate by up to 29%. Refined fabrication of NP-embedded polymer enabled the tunability of the dynamic thermal response (the ratio of internal to external dissipation rate) by a factor of 3.1 to achieve a value of 0.091, the largest reported to date. Heat dissipation rates simulated a priori were consistent with 130 μm resolution thermal images across 2- to 15-fold changes in the geometry and composition of NP-PDMS. The Nusselt number was observed to increase with the fourth root of the Rayleigh number across thermally insulative and conductive regimes, further validating the approach. These developments support the model-informed design of soft media embedded with nano-scale-spaced NPs to optimize the heat dissipation rates for evolving temperature-sensitive diagnostic and therapeutic modalities, as well as emerging uses in flexible bioelectronics, cell and tissue culture, and solar-thermal heating.
Collapse
Affiliation(s)
- D Keith Roper
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, AR 72701, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Ghaffari H, Beik J, Talebi A, Mahdavi SR, Abdollahi H. New physical approaches to treat cancer stem cells: a review. Clin Transl Oncol 2018; 20:1502-1521. [PMID: 29869042 DOI: 10.1007/s12094-018-1896-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
Cancer stem cells (CSCs) have been identified as the main center of tumor therapeutic resistance. They are highly resistant against current cancer therapy approaches particularly radiation therapy (RT). Recently, a wide spectrum of physical methods has been proposed to treat CSCs, including high energetic particles, hyperthermia (HT), nanoparticles (NPs) and combination of these approaches. In this review article, the importance and benefits of the physical CSCs therapy methods such as nanomaterial-based heat treatments and particle therapy will be highlighted.
Collapse
Affiliation(s)
- H Ghaffari
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran
| | - J Beik
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran
| | - A Talebi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran
| | - S R Mahdavi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran.
- Department of Medical Physics and Radiation Biology Research Center, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran.
| | - H Abdollahi
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Junction of Shahid Hemmat and Chamran Expressway, Tehran, Iran.
| |
Collapse
|
36
|
Wu C, Chen H, Wu X, Cong X, Wang L, Wang Y, Yang Y, Li W, Sun T. The influence of tumor-induced immune dysfunction on the immune cell distribution of gold nanoparticles in vivo. Biomater Sci 2018; 5:1531-1536. [PMID: 28589972 DOI: 10.1039/c7bm00335h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Gold nanoparticles (AuNPs) have been extensively explored as a drug carrier and have been widely used to provide advanced biomedical research tools in diagnostic imaging and therapy for cancer. Although the mononuclear phagocyte system and immune system are known to play the main roles in the clearance of AuNPs during the circulation, the particle distribution within the immune cells under the condition of immune dysfunction caused by tumor growth has not been thoroughly studied. Here, the cellular distribution of Cy5 labeled AuNPs with diameters of 5, 30 and 50 nm is characterized within the immune populations of the blood, spleen and bone marrow from tumor free and tumor bearing mice using flow cytometry. Tumor-associated immune dysfunction was observed in all immune organs and cell lineages, and it changed with tumor growth. Furthermore, the particle cellular distribution significantly changed in the tumor bearing mice compared with the tumor free mice. Finally, the particle distribution in the immune cells was also different at different stages of the tumor. Overall, these results can help inform and influence future AuNP design criteria including the future applications for nanoparticle-mediated cancer therapy.
Collapse
Affiliation(s)
- Chenxi Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Hongmei Chen
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Xuan Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Xiuxiu Cong
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Li Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yucai Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yongguang Yang
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China and Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Wei Li
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Tianmeng Sun
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
37
|
Joubert F, Pasparakis G. Hierarchically designed hybrid nanoparticles for combinational photochemotherapy against a pancreatic cancer cell line. J Mater Chem B 2018; 6:1095-1104. [PMID: 32254297 DOI: 10.1039/c7tb03261g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here, we report the formulation of hybrid nanoparticles consisting of aggregated gold nanoparticles (GNPs) impregnated into a gemcitabine-polymer conjugate matrix that exhibit synergistic photo-chemo-therapeutic activity against pancreatic cancer. Well-defined, sub-100 nm hybrid NPs were successfully formulated and their photothermal conversion efficiency was evaluated, which was found to be as high as 63% in the red-visible spectrum. By varying the GNP and GEM-polymer feed, it was possible to control the red-shifting of the surface plasmon resonance at therapeutically relevant wavelengths. The hybrid NPs exhibited significant cytotoxicity against MiaPaCa-2 cells with a half-maximal inhibitory concentration (IC50) of 0.0012 mg mL-1; however the IC50 decreased by a factor of 2 after the cells were irradiated with a continuous wave red laser for 1 min (1.4 W cm-2). Although the irradiation of the aggregated GNPs loaded in the hybrid NPs produced a higher thermal effect for the same amount of non-loaded GNPs, the IC50 of the hybrid NPs was significantly lower than that of the free GNPs, hence indicating a synergistic effect of the polymer bound GEM and the GNPs.
Collapse
Affiliation(s)
- F Joubert
- UCL School of Pharmacy, 29-39 Brunswick square, WC1N 1AX London, UK.
| | | |
Collapse
|
38
|
Yao J, Kang S, Zhang J, Du J, Zhang Z, Li M. Amphiphilic Near-Infrared Conjugated Polymer for Photothermal and Chemo Combination Therapy. ACS Biomater Sci Eng 2017; 3:2230-2234. [DOI: 10.1021/acsbiomaterials.7b00344] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jingke Yao
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
- University of the Chinese Academy of Science, Yuquan Road 19, Beijing 100049, P. R. China
| | - Shusen Kang
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
- University of the Chinese Academy of Science, Yuquan Road 19, Beijing 100049, P. R. China
| | - Jian Zhang
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
- University of the Chinese Academy of Science, Yuquan Road 19, Beijing 100049, P. R. China
| | - Jia Du
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
- University of the Chinese Academy of Science, Yuquan Road 19, Beijing 100049, P. R. China
| | - Zhe Zhang
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
| | - Mao Li
- State
Key Laboratory of Polymer Physics and Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Sciences, Remin Street
5625, Changchun 130022, P. R. China
| |
Collapse
|
39
|
A new NIR-triggered doxorubicin and photosensitizer indocyanine green co-delivery system for enhanced multidrug resistant cancer treatment through simultaneous chemo/photothermal/photodynamic therapy. Acta Biomater 2017. [PMID: 28629893 DOI: 10.1016/j.actbio.2017.06.026] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
It is a great challenge to combat multidrug resistant (MDR) cancer effectively. To address this issue, we developed a new near-infrared (NIR) triggered chemotherapeutic agent doxorubicin (DOX) and photosensitizer indocyanine green (ICG) co-release system by aid of NIR induced photothermal effect of gold nanocages (AuNCs) and temperature sensitive phase-change property of 1-tetradecanol at its melting point of 39°C, which could simultaneously exerted chemo/photothermal/photodynamic treatment on MDR human breast cancer MCF-7/ADR cells. This nano-sized system was constructed by filling the interior of AuNCs with DOX, ICG and 1-tetradecanol, and modifying the surface with biotinylated poly (ethylene glycol) via Au-S bonds, termed as DOX/ICG@biotin-PEG-AuNC-PCM. The DOX and ICG co-release from DOX/ICG@biotin-PEG-AuNC-PCM was much faster in PBS at 40°C or under 808nm NIR irradiation at 2.5W/cm2 than at 37°C (e.g. 67.27% or 80.31% vs. 5.57% of DOX, 76.08% vs. 3.83% of ICG for 20min). The flow cytometry and confocal laser scanning microscopy (CLSM) results showed, the AuNCs were taken up by MCF-7/ADR cells via endocytosis, thus enhancing DOX uptake; the biotin on AuNCs facilitated this endocytosis; NIR irradiation caused the heating of the AuNCs, triggering the DOX and ICG co-release and enhancing the distribution of DOX in nuclei, the released ICG generated ROS to take photodynamic therapy. Due to the above unique properties, DOX/ICG@biotin-PEG-AuNC-PCM exerted excellent anti-tumor effects under NIR irradiation, its IC50 against MCF-7/ADR cells was very low, only 0.48µg/mL, much smaller than that of free DOX (74.51μg/mL). STATEMENT OF SIGNIFICANCE A new near-infrared (NIR) triggered chemotherapeutic agent doxorubicin (DOX) and photosensitizer indocyanine green (ICG) co-release system by aid of NIR induced photothermal effect of gold nanocages (AuNCs) and temperature sensitive phase-change property of 1-tetradecanol at its melting point of 39°C, was prepared, termed as DOX/ICG@biotin-PEG-AuNC-PCM, which could simultaneously exerted chemo/photothermal/photodynamic treatment on MDR human breast cancer MCF-7/ADR cells. DOX/ICG@biotin-PEG-AuNC-PCM exerted excellent anti-tumor effects under NIR irradiation, its IC50 against MCF-7/ADR cells was very low, only 0.48µg/mL, much smaller than that of free DOX (74.51μg/mL).
Collapse
|
40
|
Tang J, Zhou H, Liu J, Liu J, Li W, Wang Y, Hu F, Huo Q, Li J, Liu Y, Chen C. Dual-Mode Imaging-Guided Synergistic Chemo- and Magnetohyperthermia Therapy in a Versatile Nanoplatform To Eliminate Cancer Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:23497-23507. [PMID: 28661121 DOI: 10.1021/acsami.7b06393] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Cancer stem cells (CSCs) have been identified as a new target for therapy in diverse cancers. Traditional therapies usually kill the bulk of cancer cells, but are often unable to effectively eliminate CSCs, which may lead to drug resistance and cancer relapse. Herein, we propose a novel strategy: fabricating multifunctional magnetic Fe3O4@PPr@HA hybrid nanoparticles and loading it with the Notch signaling pathway inhibitor N-[N-(3,5-difluorophenacetyl-l-alanyl)]-S-phenylglycinet-butylester (DAPT) to eliminate CSCs. Hyaluronic acid ligands greatly enhance the accumulation of the hybrid nanoparticles in the tumor site and in the CSCs. Both hyaluronase in the tumor microenvironment and the magnetic hyperthermia effect of the inner magnetic core can accelerate the release of DAPT. This controlled release of DAPT in the tumor site further enhances the ability of the combination of chemo- and magnetohyperthermia therapy to eliminate cancer stem cells. With the help of polypyrrole-mediated photoacoustic and Fe3O4-mediated magnetic resonance imaging, the drug release can be precisely monitored in vivo. This versatile nanoplatform enables effective elimination of the cancer stem cells and monitoring of the drugs.
Collapse
Affiliation(s)
- Jinglong Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jiaming Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wanqi Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
| | - Yuqing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
| | - Fan Hu
- Department of Biomedical, College of Biochemical Engineering, Beijing Union University , Beijing 100023, China
| | - Qing Huo
- Department of Biomedical, College of Biochemical Engineering, Beijing Union University , Beijing 100023, China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
41
|
Cao Y, Yi J, Yang X, Liu L, Yu C, Huang Y, Sun L, Bao Y, Li Y. Efficient Cancer Regression by a Thermosensitive Liposome for Photoacoustic Imaging-Guided Photothermal/Chemo Combinatorial Therapy. Biomacromolecules 2017; 18:2306-2314. [DOI: 10.1021/acs.biomac.7b00464] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yue Cao
- National
Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117 Jilin, China
| | - Jingwen Yi
- National
Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117 Jilin, China
| | - Xiaoguang Yang
- National
Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117 Jilin, China
| | - Lei Liu
- National
Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117 Jilin, China
| | | | | | | | | | - Yuxin Li
- National
Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun 130117 Jilin, China
| |
Collapse
|
42
|
Shen S, Zhu C, Huo D, Yang M, Xue J, Xia Y. A Hybrid Nanomaterial for the Controlled Generation of Free Radicals and Oxidative Destruction of Hypoxic Cancer Cells. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201702898] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Song Shen
- The Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology and Emory University; Atlanta GA 30332 USA
- College of Pharmaceutical Sciences; Jiangsu University; Zhenjiang Jiangsu 212013 China
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology and Emory University; Atlanta GA 30332 USA
| | - Da Huo
- The Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology and Emory University; Atlanta GA 30332 USA
| | - Miaoxin Yang
- School of Chemistry and Biochemistry; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology and Emory University; Atlanta GA 30332 USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology and Emory University; Atlanta GA 30332 USA
- School of Chemistry and Biochemistry; Georgia Institute of Technology; Atlanta GA 30332 USA
| |
Collapse
|
43
|
Shen S, Zhu C, Huo D, Yang M, Xue J, Xia Y. A Hybrid Nanomaterial for the Controlled Generation of Free Radicals and Oxidative Destruction of Hypoxic Cancer Cells. Angew Chem Int Ed Engl 2017; 56:8801-8804. [PMID: 28464414 DOI: 10.1002/anie.201702898] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Indexed: 01/16/2023]
Abstract
Anticancer modalities based on oxygen free radicals, including photodynamic therapy and radiotherapy, have emerged as promising treatments in the clinic. However, the hypoxic environment in tumor tissue prevents the formation of oxygen free radicals. Here we introduce a novel strategy that employs oxygen-independent free radicals generated from a polymerization initiator for eradicating cancer cells. The initiator is mixed with a phase-change material and loaded into the cavities of gold nanocages. Upon irradiation by a near-infrared laser, the phase-change material is melted due to the photothermal effect of gold nanocages, leading to the release and decomposition of the loaded initiator to generate free radicals. The free radicals produced in this way are highly effective in inducing apoptosis in hypoxic cancer cells.
Collapse
Affiliation(s)
- Song Shen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.,College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Da Huo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Miaoxin Yang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA.,School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
44
|
Yang Y, Aw J, Xing B. Nanostructures for NIR light-controlled therapies. NANOSCALE 2017; 9:3698-3718. [PMID: 28272614 DOI: 10.1039/c6nr09177f] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In general, effective clinical treatment demands precision medicine, which requires specific perturbation to disease cells with no damage to normal tissue. Thus far, guaranteeing that selective therapeutic effects occur only at targeted disease areas remains a technical challenge. Among the various endeavors to achieve such an outcome, strategies based on light-controlled therapies have received special attention, mostly due to their unique advantages, including the low-invasive property and the capability to obtain spatial and temporal precision at the targeted sites via specific wavelength light irradiation. However, most conventional light-mediated therapies, especially those based on short-wavelength UV or visible light irradiation, have potential issues including limited penetration depth and harmful photo damage to healthy tissue. Therefore, the implemention of near-infrared (NIR) light illumination, which can travel into deeper tissues without causing obvious photo-induced cytotoxcity, has been suggested as a preferable option for precise phototherapeutic applications in vitro and in vivo. In this article, an overview is presented of existing therapeutic applications through NIR light-absorbed nanostructures, such as NIR light-controlled drug delivery, NIR light-mediated photothermal and photodynamic therapies. Potential challenges and relevant future prospects are also discussed.
Collapse
Affiliation(s)
- Yanmei Yang
- Center for Molecular Imaging and Nuclear Medicine, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China 215123.
| | - Junxin Aw
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, 637371, Singapore and Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore, 117602, Singapore
| |
Collapse
|
45
|
Abstract
A complex PGRMC1-centred regulatory system controls multiple cell functions. Although PGRMC1 is phosphorylated at several positions, we do not understand the mechanisms regulating its function. PGRMC1 is the archetypal member of the membrane associated progesterone receptor (MAPR) family. Phylogentic comparison of MAPR proteins suggests that the ancestral metazoan "PGRMC-like" MAPR gene resembled PGRMC1/PGRMC2, containing the equivalents of PGRMC1 Y139 and Y180 SH2 target motifs. It later acquired a CK2 site with phosphoacceptor at S181. Separate PGRMC1 and PGRMC2 genes with this "PGRMC-like" structure diverged after the separation of vertebrates from protochordates. Terrestrial tetrapods possess a novel proline-rich PGRMC1 SH3 target motif centred on P64 which in mammals is augmented by a phosphoacceptor at PGRMC1 S54, and in primates by an additional S57 CK2 site. All of these phosphoacceptors are phosphorylated in vivo. This study suggests that an increasingly sophisticated system of PGRMC1-modulated multicellular functional regulation has characterised animal evolution since Precambrian times.
Collapse
|
46
|
Qin W, Huang G, Chen Z, Zhang Y. Nanomaterials in Targeting Cancer Stem Cells for Cancer Therapy. Front Pharmacol 2017; 8:1. [PMID: 28149278 PMCID: PMC5241315 DOI: 10.3389/fphar.2017.00001] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/03/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified in almost all cancers and give rise to metastases and can also act as a reservoir of cancer cells that may cause a relapse after surgery, radiation, or chemotherapy. Thus they are obvious targets in therapeutic approaches and also a great challenge in cancer treatment. The threat presented by CSCs lies in their unlimited proliferative ability and multidrug resistance. These findings have necessitated an effective novel strategy to target CSCs for cancer treatment. Nanomaterials are on the route to providing novel methods in cancer therapies. Although, there have been a large number of excellent work in the field of targeted cancer therapy, it remains an open question how nanomaterials can meet future demands for targeting and eradicating of CSCs. In this review, we summarized recent and highlighted future prospects for targeting CSCs for cancer therapies by using a variety of nanomaterials.
Collapse
Affiliation(s)
- Weiwei Qin
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Guan Huang
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Zuanguang Chen
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Yuanqing Zhang
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| |
Collapse
|
47
|
The Evolution of the Sigma-2 (σ 2) Receptor from Obscure Binding Site to Bona Fide Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:49-61. [PMID: 28315264 DOI: 10.1007/978-3-319-50174-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sigma-2 (σ2) receptor represents one of the most poorly understood proteins in cell biology. Although this receptor was identified through in vitro binding studies over 25 years ago, the molecular identity of this protein is currently not unambiguously known, and the results from recent attempts to identify the σ2 receptor through protein purification and mass spectral analysis have been the subject of debate in the literature. However, there is overwhelming data demonstrating that the σ2 receptor is an important biomarker of tumor cell proliferation . The observation that σ2 receptor agonists are potent anticancer agents whereas σ2 antagonists block Aβ1-42 oligomer synaptic dysfunction in transgenic mouse models of Alzheimer's disease have clearly identified this protein as an important therapeutic target for the treatment of a variety of pathological conditions.
Collapse
|
48
|
Wang S, Lin J, Wang T, Chen X, Huang P. Recent Advances in Photoacoustic Imaging for Deep-Tissue Biomedical Applications. Theranostics 2016; 6:2394-2413. [PMID: 27877243 PMCID: PMC5118603 DOI: 10.7150/thno.16715] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
Photoacoustic imaging (PAI), a novel imaging modality based on photoacoustic effect, shows great promise in biomedical applications. By converting pulsed laser excitation into ultrasonic emission, PAI combines the advantages of optical imaging and ultrasound imaging, which benefits rich contrast, high resolution and deep tissue penetration. In this paper, we introduced recent advances of contrast agents, applications, and signal enhancement strategies for PAI. The PA contrast agents were categorized by their components, mainly including inorganic and organic PA contrast agents. The applications of PAI were summarized as follows: deep tumor imaging, therapeutic responses monitoring, metabolic imaging, pH detection, enzyme detection, reactive oxygen species (ROS) detection, metal ions detection, and so on. The enhancement strategies of PA signals were highlighted. In the end, we elaborated on the challenges and provided perspectives of PAI for deep-tissue biomedical applications.
Collapse
Affiliation(s)
- Sheng Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jing Lin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
49
|
He C, Poon C, Chan C, Yamada SD, Lin W. Nanoscale Coordination Polymers Codeliver Chemotherapeutics and siRNAs to Eradicate Tumors of Cisplatin-Resistant Ovarian Cancer. J Am Chem Soc 2016; 138:6010-9. [PMID: 27088560 DOI: 10.1021/jacs.6b02486] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug resistance impedes the successful treatment of many types of cancers, especially ovarian cancer (OCa). To counter this problem, we developed novel long-circulating, self-assembled core-shell nanoscale coordination polymer (NCP) nanoparticles that efficiently deliver multiple therapeutics with different mechanisms of action to enhance synergistic therapeutic effects. These NCP particles contain high payloads of chemotherapeutics cisplatin or cisplatin plus gemcitabine in the core and pooled siRNAs that target multidrug resistant (MDR) genes in the shell. The NCP particles possess efficient endosomal escape via a novel carbon dioxide release mechanism without compromising the neutral surface charge required for long blood circulation and effectively downregulate MDR gene expression in vivo to enhance chemotherapeutic efficacy by several orders of magnitude. Even at low doses, intraperitoneal injections of nanoparticles led to effective and long-lasting tumor regression/eradication in subcutaneous and intraperitoneal xenograft mouse models of cisplatin-resistant OCa. By silencing MDR genes in tumors, self-assembled core-shell nanoparticles promise a more effective chemotherapeutic treatment for many challenging cancers.
Collapse
Affiliation(s)
- Chunbai He
- Department of Chemistry, The University of Chicago , Chicago, Illinois 60637, United States
| | - Christopher Poon
- Department of Chemistry, The University of Chicago , Chicago, Illinois 60637, United States
| | - Christina Chan
- Department of Chemistry, The University of Chicago , Chicago, Illinois 60637, United States
| | - S Diane Yamada
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, The University of Chicago , Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago , Chicago, Illinois 60637, United States
| |
Collapse
|
50
|
Zou L, Wang H, He B, Zeng L, Tan T, Cao H, He X, Zhang Z, Guo S, Li Y. Current Approaches of Photothermal Therapy in Treating Cancer Metastasis with Nanotherapeutics. Theranostics 2016; 6:762-72. [PMID: 27162548 PMCID: PMC4860886 DOI: 10.7150/thno.14988] [Citation(s) in RCA: 590] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/06/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer metastasis accounts for the high mortality of many types of cancer. Owing to the unique advantages of high specificity and minimal invasiveness, photothermal therapy (PTT) has been evidenced with great potential in treating cancer metastasis. In this review, we outline the current approaches of PTT with respect to its application in treating metastatic cancer. PTT can be used alone, guided with multimodal imaging, or combined with the current available therapies for effective treatment of cancer metastasis. Numerous types of photothermal nanotherapeutics (PTN) have been developed with encouraging therapeutic efficacy on metastatic cancer in many preclinical animal experiments. We summarize the design and performance of various PTN in PTT alone and their combinational therapy. We also point out the lacking area and the most promising approaches in this challenging field. In conclusion, PTT or their combinational therapy can provide an essential promising therapeutic modality against cancer metastasis.
Collapse
|