1
|
Xu Y, Dang H, Teng C, Yin D, Yan L. ATP Inhibition for Starvation/Mild Photothermal/Photodynamic Synergy Therapy Using Polypeptide Nanoparticles Conjugating 2-Deoxy-D-Glucose and Dye under NIR Phototheranostic Strategy. Adv Healthc Mater 2024; 13:e2401219. [PMID: 38758576 DOI: 10.1002/adhm.202401219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/27/2024] [Indexed: 05/18/2024]
Abstract
Rapid propagation of tumor cells requires plenty of energy, which is adenosine triphosphate (ATP) dependent. ATP inhibition in tumors not only results in the starvation of tumor cells but also down-regulation of the level of heat shock proteins (HSPs), which usually increase during traditional photothermal therapy (PTT), especially when the temperature is up 50 °C. 2-deoxy-D-glucose (2DG) is an anti-glycolytic reagent and can be used as an efficient agent for ATP inhibition in tumors. Compared with typical PTT, low-temperature mild photothermal therapy (MPTT) is receiving more and more attention because it avoids the high temperatures causing damage to the normal tissue, and the increase of HSPs which decrease PTT. Here, multifunctional polypeptide nanoparticles pDG@Ahx conjugating both a NIR probe Ahx-BDP and 2DG into the side chain of the amphiphilic polypeptide have been prepared. In vitro and in vivo studies reveal that the as-prepared nanoparticles achieve a synergistic effect of starvation/MPTT/PDT (photodynamic therapy), and it provides a new strategy to NIR-I/II fluorescence imaging-guided starvation/MPTT/PDT synergy therapy for tumors.
Collapse
Affiliation(s)
- Yixuan Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Huiping Dang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Changchang Teng
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Dalong Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
| | - Lifeng Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| |
Collapse
|
2
|
Shen J, Chen G, Zhao L, Huang G, Liu H, Liu B, Miao Y, Li Y. Recent Advances in Nanoplatform Construction Strategy for Alleviating Tumor Hypoxia. Adv Healthc Mater 2023; 12:e2300089. [PMID: 37055912 DOI: 10.1002/adhm.202300089] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Hypoxia is a typical feature of most solid tumors and has important effects on tumor cells' proliferation, invasion, and metastasis. This is the key factor that leads to poor efficacy of different kinds of therapy including chemotherapy, radiotherapy, photodynamic therapy, etc. In recent years, the construction of hypoxia-relieving functional nanoplatforms through nanotechnology has become a new strategy to reverse the current situation of tumor microenvironment hypoxia and improve the effectiveness of tumor treatment. Here, the main strategies and recent progress in constructing nanoplatforms are focused on to directly carry oxygen, generate oxygen in situ, inhibit mitochondrial respiration, and enhance blood perfusion to alleviate tumor hypoxia. The advantages and disadvantages of these nanoplatforms are compared. Meanwhile, nanoplatforms based on organic and inorganic substances are also summarized and classified. Through the comprehensive overview, it is hoped that the summary of these nanoplatforms for alleviating hypoxia could provide new enlightenment and prospects for the construction of nanomaterials in this field.
Collapse
Affiliation(s)
- Jing Shen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guobo Chen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Linghao Zhao
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Hui Liu
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
3
|
Vakhrushev AV, Gruzdev DA, Demin AM, Levit GL, Krasnov VP. Synthesis of Novel Carborane-Containing Derivatives of RGD Peptide. Molecules 2023; 28:molecules28083467. [PMID: 37110700 PMCID: PMC10143838 DOI: 10.3390/molecules28083467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Short peptides containing the Arg-Gly-Asp (RGD) fragment can selectively bind to integrins on the surface of tumor cells and are attractive transport molecules for the targeted delivery of therapeutic and diagnostic agents to tumors (for example, glioblastoma). We have demonstrated the possibility of obtaining the N- and C-protected RGD peptide containing 3-amino-closo-carborane and a glutaric acid residue as a linker fragment. The resulting carboranyl derivatives of the protected RGD peptide are of interest as starting compounds in the synthesis of unprotected or selectively protected peptides, as well as building blocks for preparation of boron-containing derivatives of the RGD peptide of a more complex structure.
Collapse
Affiliation(s)
- Alexander V Vakhrushev
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 620108 Ekaterinburg, Russia
| | - Dmitry A Gruzdev
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 620108 Ekaterinburg, Russia
| | - Alexander M Demin
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 620108 Ekaterinburg, Russia
| | - Galina L Levit
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 620108 Ekaterinburg, Russia
| | - Victor P Krasnov
- Postovsky Institute of Organic Synthesis, Russian Academy of Sciences (Ural Branch), 620108 Ekaterinburg, Russia
| |
Collapse
|
4
|
Zhang L, Liu X, Mao Y, Rong S, Chen Y, Qi Y, Cai Z, Li H. Inhibition of melanoma using a nanoceria-based prolonged oxygen-generating phototherapy hydrogel. Front Oncol 2023; 13:1126094. [PMID: 37007107 PMCID: PMC10060878 DOI: 10.3389/fonc.2023.1126094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/23/2023] [Indexed: 03/18/2023] Open
Abstract
Tumor hypoxic environment is an inevitable obstacle for photodynamic therapy (PDT) of melanoma. Herein, a multifunctional oxygen-generating hydrogel loaded with hyaluronic acid-chlorin e6 modified nanoceria and calcium peroxide (Gel-HCeC-CaO2) was developed for the phototherapy of melanoma. The thermo-sensitive hydrogel could act as a sustained drug delivery system to accumulate photosensitizers (chlorin e6, Ce6) around the tumor, followed by cellular uptake mediated by nanocarrier and hyaluronic acid (HA) targeting. The moderate sustained oxygen generation in the hydrogel was produced by the reaction of calcium peroxide (CaO2) with infiltrated H2O in the presence of catalase mimetic nanoceria. The developed Gel-HCeC-CaO2 could efficiently alleviate the hypoxia microenvironment of tumors as indicated by the expression of hypoxia-inducible factor -1α (HIF-1α), meeting the “once injection, repeat irradiation” strategy and enhanced PDT efficacy. The prolonged oxygen-generating phototherapy hydrogel system provided a new strategy for tumor hypoxia alleviation and PDT.
Collapse
Affiliation(s)
- Lidong Zhang
- Institute of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaoguang Liu
- Department of Gynecology, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yinghua Mao
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing, China
| | - Shu Rong
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing, China
| | - Yonghong Chen
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing, China
| | - Yong Qi
- Huadong Research Institute for Medicine and Biotechniques, Nanjing, China
| | - Zhipeng Cai
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing, China
| | - Hong Li
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing, China
- *Correspondence: Hong Li,
| |
Collapse
|
5
|
Tumor Spheroids as Model to Design Acoustically Mediated Drug Therapies: A Review. Pharmaceutics 2023; 15:pharmaceutics15030806. [PMID: 36986667 PMCID: PMC10056013 DOI: 10.3390/pharmaceutics15030806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Tumor spheroids as well as multicellular tumor spheroids (MCTSs) are promising 3D in vitro tumor models for drug screening, drug design, drug targeting, drug toxicity, and validation of drug delivery methods. These models partly reflect the tridimensional architecture of tumors, their heterogeneity and their microenvironment, which can alter the intratumoral biodistribution, pharmacokinetics, and pharmacodynamics of drugs. The present review first focuses on current spheroid formation methods and then on in vitro investigations exploiting spheroids and MCTS for designing and validating acoustically mediated drug therapies. We discuss the limitations of the current studies and future perspectives. Various spheroid formation methods enable the easy and reproducible generation of spheroids and MCTSs. The development and assessment of acoustically mediated drug therapies have been mainly demonstrated in spheroids made up of tumor cells only. Despite the promising results obtained with these spheroids, the successful evaluation of these therapies will need to be addressed in more relevant 3D vascular MCTS models using MCTS-on-chip platforms. These MTCSs will be generated from patient-derived cancer cells and nontumor cells, such as fibroblasts, adipocytes, and immune cells.
Collapse
|
6
|
Oxygen-carrying nanoplatform to reprogram tumor immunosuppressive microenvironment and enhance photothermal-immunotherapy. Mater Today Bio 2023; 19:100555. [PMID: 36793322 PMCID: PMC9922928 DOI: 10.1016/j.mtbio.2023.100555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/19/2023] Open
Abstract
Immunotherapy shows great promise on treating tumors. However, insufficient antigen exposure and immunosuppressive tumor microenvironment (TME) caused by hypoxia impose a serial of constraints on the therapeutic efficacy. In this study, we developed an oxygen-carrying nanoplatform loaded with perfluorooctyl bromide (PFOB, a second-generation of perfluorocarbon-based blood substitute), IR780 (a photosensitizer) and imiquimod (R837, an immune adjuvant) to reprogram immunosuppressive TME and reinforce photothermal-immunotherapy. The obtained oxygen-carrying nanoplatforms (abbreviated as IR-R@LIP/PFOB) show highly efficient oxygen release behavior and excellent hyperthermia performance upon laser irradiation, thus achieving the attenuation of the inherent tumor hypoxia and the exposure of tumor associated antigens in situ, and transforming the immunosuppressive TME to an immunosupportive one. We found that the photothermal therapy of IR-R@LIP/PFOB together with anti-programmed cell death protein-1 (anti-PD-1) would elicit a robust antitumor immunity by increasing the tumor-infiltrating frequencies of cytotoxic CD8+ T cells and tumoricidal M1-phenotype macrophages, while reducing immunosuppressive M2-phenotype macrophages and regulatory T cells (Tregs). This study presents these oxygen-carrying IR-R@LIP/PFOB nanoplatforms are potent in removing some negative impacts of immunosuppressive TME caused by hypoxia, and suppressing tumor growth by initiating antitumor immune responses, especially in combination with anti-PD-1 immunotherapy.
Collapse
|
7
|
Mosqueira VCF, Machado MGC, de Oliveira MA. Polymeric Nanocarriers in Cancer Theranostics. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
8
|
Wang D, Li H, Wang D, Hao Y, Gui H, Liu J, Zhang Y, Liu J, Yang C. Supramolecular Coassembled Peptide Hydrogels for Efficient Anticancer Therapy by RNS-Based PDT and Immune Microenvironment Regulation. Macromol Biosci 2022; 22:e2200359. [PMID: 36208072 DOI: 10.1002/mabi.202200359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Indexed: 01/15/2023]
Abstract
Photodynamic therapy (PDT) has attracted much attention in cancer treatment due to its tumor selectivity and noninvasive nature. Recent studies have demonstrated that PDT mediated reactive oxygen species (ROS) generation in tumor microenvironment (TME) synergistically improves the efficacy of immune checkpoint blockade (ICB) therapy. However, the instability and short half-life of the ROS generated by PDT limit its clinical applications. Herein, a coassembled peptide hydrogel comprising two short peptides that contained the same assembly unit, Ce6-KKFKFEFEF (KEF-Ce6) and RRRRRRRR-KFKFEFEF (KEF-R8) is developed. When exposed to 635 nm laser irradiation, KEF-Ce6 released ROS, while KEF-R8 plays as nitric oxide (NO) donor. Subsequently, ROS reacts with NO to produce reactive nitrogen species (RNS). Both in vitro and in vivo experiments prove that converting ROS into more cytotoxic RNS caused intense cell death. Importantly, it is observed that tumor-associated macrophages (TAMs) are polarized to proinflammatory types (M1-type) by the RNS-based PDT. The increase of M1 macrophages relieves the immunosuppressive situation in TME. Thus, when combined with αPD-L1 treatment, the survival time of tumor-bearing mice is prolonged. Overall, a simple yet efficient coassembled hydrogel that can cascade release ROS/NO/RNS and strengthen antitumor T cell responses to boost cancer immunotherapy by reprogramming TAMs is provided.
Collapse
Affiliation(s)
- Dan Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Hui Li
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Dianyu Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Yusen Hao
- Lab of Functional and Biomedical Nanomaterials College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Han Gui
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
9
|
Alves CG, Lima-Sousa R, Melo BL, Ferreira P, Moreira AF, Correia IJ, Melo-Diogo DD. Poly(2-ethyl-2-oxazoline)-IR780 conjugate nanoparticles for breast cancer phototherapy. Nanomedicine (Lond) 2022; 17:2057-2072. [PMID: 36803049 DOI: 10.2217/nnm-2022-0218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Aims: To address the limitations of IR780 by preparing hydrophilic polymer-IR780 conjugates and to employ these conjugates in the assembly of nanoparticles (NPs) intended for cancer photothermal therapy. Materials & methods: The cyclohexenyl ring of IR780 was conjugated for the first time with thiol-terminated poly(2-ethyl-2-oxazoline) (PEtOx). This novel poly(2-ethyl-2-oxazoline)-IR780 (PEtOx-IR) conjugate was combined with D-α-tocopheryl succinate (TOS), leading to the assembly of mixed NPs (PEtOx-IR/TOS NPs). Results: PEtOx-IR/TOS NPs displayed optimal colloidal stability as well as cytocompatibility in healthy cells at doses within the therapeutic range. In turn, the combination of PEtOx-IR/TOS NPs and near-infrared light reduced heterotypic breast cancer spheroid viability to just 15%. Conclusion: PEtOx-IR/TOS NPs are promising agents for breast cancer photothermal therapy.
Collapse
Affiliation(s)
- Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
| | - Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
| | - Bruna L Melo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
| | - Paula Ferreira
- CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, Coimbra, 3030-790, Portugal
- Department of Chemical & Biological Engineering, Coimbra Institute of Engineering (ISEC), Rua Pedro Nunes, Coimbra, 3030-199, Portugal
| | - André F Moreira
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
- CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, Coimbra, 3030-790, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Avenida Infante D Henrique, Covilhã, 6200-506, Portugal
| |
Collapse
|
10
|
Xu J, Ning J, Wang Y, Xu M, Yi C, Yan F. Carbon dots as a promising therapeutic approach for combating cancer. Bioorg Med Chem 2022; 72:116987. [DOI: 10.1016/j.bmc.2022.116987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/08/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022]
|
11
|
Menilli L, Milani C, Reddi E, Moret F. Overview of Nanoparticle-Based Approaches for the Combination of Photodynamic Therapy (PDT) and Chemotherapy at the Preclinical Stage. Cancers (Basel) 2022; 14:cancers14184462. [PMID: 36139623 PMCID: PMC9496990 DOI: 10.3390/cancers14184462] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The present review represents the outstanding and promising recent literature reports (2017–2022) on nanoparticle-based formulations developed for anticancer therapy with photodynamic therapy (PDT), photosensitizers, and chemotherapeutics. Besides brief descriptions of chemotherapeutics’ classification and of PDT mechanisms and limitations, several examples of nanosystems endowed with different responsiveness (e.g., acidic pH and reactive oxygen species) and peculiarity (e.g., tumor oxygenation capacity, active tumor targeting, and biomimetic features) are described, and for each drug combination, in vitro and in vivo results on preclinical cancer models are reported. Abstract The widespread diffusion of photodynamic therapy (PDT) as a clinical treatment for solid tumors is mainly limited by the patient’s adverse reaction (skin photosensivity), insufficient light penetration in deeply seated neoplastic lesions, unfavorable photosensitizers (PSs) biodistribution, and photokilling efficiency due to PS aggregation in biological environments. Despite this, recent preclinical studies reported on successful combinatorial regimes of PSs with chemotherapeutics obtained through the drugs encapsulation in multifunctional nanometric delivery systems. The aim of the present review deals with the punctual description of several nanosystems designed not only with the objective of co-transporting a PS and a chemodrug for combination therapy, but also with the goal of improving the therapeutic efficacy by facing the main critical issues of both therapies (side effects, scarce tumor oxygenation and light penetration, premature drug clearance, unspecific biodistribution, etc.). Therefore, particular attention is paid to the description of bio-responsive drugs and nanoparticles (NPs), targeted nanosystems, biomimetic approaches, and upconverting NPs, including analyzing the therapeutic efficacy of the proposed photo-chemotherapeutic regimens in in vitro and in vivo cancer models.
Collapse
Affiliation(s)
- Luca Menilli
- Department of Biology, University of Padova, 35100 Padova, Italy
| | - Celeste Milani
- Department of Biology, University of Padova, 35100 Padova, Italy
- Institute of Organic Synthesis and Photoreactivity, ISOF-CNR, 40129 Bologna, Italy
| | - Elena Reddi
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| | - Francesca Moret
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| |
Collapse
|
12
|
Luo Y, Qiao B, Yang C, Zhang P, Xie Z, Cao J, Yang A, Xiang Q, Ran H, Wang Z, Hao L, Cao Y, Zhou Z, Ren J. Low Intensity Focused Ultrasound Ignited “Deep-Penetration Nanobomb” (DPNB) for Tetramodal Imaging Guided Hypoxia-Tolerant Sonodynamic Therapy Against Hypoxic Tumors. Int J Nanomedicine 2022; 17:4547-4565. [PMID: 36199475 PMCID: PMC9527552 DOI: 10.2147/ijn.s361648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/31/2022] [Indexed: 12/07/2022] Open
Abstract
Background Sonodynamic therapy (SDT) has been regarded as a novel therapeutic modality for killing tumors. However, the hypoxic tumor microenvironment, especially deep-seated tumors distant from blood vessels, severely restricts therapeutic efficacy due to the oxygen-dependent manner of SDT. Methods Herein, we report a novel ultrasonic cavitation effect-based therapeutic modality that is able to facilitate the hypoxia-tolerant SDT for inducing hypoxic tumor death. A tLyP-1 functionalized liposomes is fabricated, composed of hematoporphyrin monomethyl ether gadolinium as the sonosentizer and perfluoropentane (PFP) as the acoustic environment regulator. Moreover, the tLyP-1 functioned liposomes could achieve active tumor homing and effective deep-penetrating into hypoxic tumors. Upon low intensity focused ultrasound (LIFU) irradiation, the acoustic droplet vaporization effect of PFP induced fast liquid-to-gas transition and quick bubbles explosion to generate hydroxyl radicals, efficiently promoting cell death in both normoxic and hypoxic microenvironment (acting as deep-penetration nanobomb, DPNB). Results The loading of PFP is proved to significantly enhance the therapeutic efficacy of hypoxic tumors. In particular, these DPNB can also act as ultrasound, photoacoustic, magnetic resonance, and near-infrared fluorescence tetramodal imaging agents for guiding the therapeutic process. Conclusion This study is the first report involving that liquid-to-gas transition based SDT has the potential to combat hypoxic tumors.
Collapse
Affiliation(s)
- Yuanli Luo
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Bin Qiao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Chao Yang
- Radiology Department, Chongqing General Hospital, Chongqing, 400014, People’s Republic of China
| | - Ping Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Zhuoyan Xie
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Jin Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Anyu Yang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Qinyanqiu Xiang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Lan Hao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Zhiyi Zhou
- General Practice Department, Chongqing General Hospital, Chongqing, 400014, People’s Republic of China
- Correspondence: Zhiyi Zhou; Jianli Ren, Email ;
| | - Jianli Ren
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| |
Collapse
|
13
|
Zhang Y, Li K, Han X, Chen Q, Shao L, Bai D. A photochemical-responsive nanoparticle boosts doxorubicin uptake to suppress breast cancer cell proliferation by apoptosis. Sci Rep 2022; 12:10354. [PMID: 35725767 PMCID: PMC9209492 DOI: 10.1038/s41598-022-14518-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023] Open
Abstract
In the course of chemotherapy for breast cancer, doxorubicin (DOX) is one of the most commonly prescribed agents. However, it has been recognized as clinically circumscribed on account of its poor selectivity and toxic reactions to normal tissues. Fortunately, the distinct merit of photochemical-responsive nanoparticle delivery systems to enhance cellular drugs uptake through localized concentration, adequate selective and minimizing systemic toxicity has aroused substantial interest recently. In this study, we synthesized photochemical-responsive nanoparticle by incorporating DOX, curcumin (CUR), and perfluorooctyl bromide (PFOB) into poly(lactic-co-glycolic acid) (PLGA) via double emulsification (DOX-CUR-PFOB-PLGA). The synthesized composite nanoparticles, which featured good ultrasound imaging, engendered photochemical activation for drug release when given laser irradiation. Cumulative release rates for DOX were 76.34%, and for CUR were 83.64%, respectively. Also, MCF-7 cells displayed significant intracellular DOX uptake and reactive oxygen species (ROS) levels, degraded cytoskeleton, and decreased cell growth and migration capacity. At the molecular level, cellular pAKT levels decreased, which resulted in downregulated HIF-1α and BAX/BCl-2 levels, leading to Caspase-3 activation and thus induction of apoptosis. Therefore, the photochemical-responsive nanoparticles possess the potential to elicit apoptosis in MCF-7 cells via enhanced DOX uptake.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kaiting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyu Han
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qing Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lan Shao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
14
|
Sivasubramanian M, Lo LW. Assessment of Nanoparticle-Mediated Tumor Oxygen Modulation by Photoacoustic Imaging. BIOSENSORS 2022; 12:336. [PMID: 35624636 PMCID: PMC9138624 DOI: 10.3390/bios12050336] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 06/01/2023]
Abstract
Photoacoustic imaging (PAI) is an invaluable tool in biomedical imaging, as it provides anatomical and functional information in real time. Its ability to image at clinically relevant depths with high spatial resolution using endogenous tissues as contrast agents constitutes its major advantage. One of the most important applications of PAI is to quantify tissue oxygen saturation by measuring the differential absorption characteristics of oxy and deoxy Hb. Consequently, PAI can be utilized to monitor tumor-related hypoxia, which is a crucial factor in tumor microenvironments that has a strong influence on tumor invasiveness. Reactive oxygen species (ROS)-based therapies, such as photodynamic therapy, radiotherapy, and sonodynamic therapy, are oxygen-consuming, and tumor hypoxia is detrimental to their efficacy. Therefore, a persistent demand exists for agents that can supply oxygen to tumors for better ROS-based therapeutic outcomes. Among the various strategies, NP-mediated supplemental tumor oxygenation is especially encouraging due to its physio-chemical, tumor targeting, and theranostic properties. Here, we focus on NP-based tumor oxygenation, which includes NP as oxygen carriers and oxygen-generating strategies to alleviate hypoxia monitored by PAI. The information obtained from quantitative tumor oxygenation by PAI not only supports optimal therapeutic design but also serves as a highly effective tool to predict therapeutic outcomes.
Collapse
Affiliation(s)
| | - Leu-Wei Lo
- Department of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 350, Taiwan;
| |
Collapse
|
15
|
Homologous targeting nanoparticles for enhanced PDT against osteosarcoma HOS cells and the related molecular mechanisms. J Nanobiotechnology 2022; 20:83. [PMID: 35177075 PMCID: PMC8851855 DOI: 10.1186/s12951-021-01201-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND No prominent advancements in osteosarcoma (OS) treatment have been made in the past 20 years. Although photodynamic therapy (PDT) is an emerging technique for cancer therapy, the lack of targeted photosensitizers for OS treatment severely limits its applications. RESULTS In this study, we constructed a potential theranostic nanoplatform by using (poly (lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) encapsulating IR780 into the shell (PLGA-IR780 NPs), which were further camouflaged with human OS cell membranes from the HOS cell line (MH-PLGA-IR780 NPs). These constructed NPs showed the capacity for homologous targeting with excellent photoacoustic (PA)/fluorescence (FL) imaging ability. Benefitting from their homologous targeting capacity, MH-PLGA-IR780 NPs obviously promoted cell endocytosis in vitro and tumor accumulation in vivo, which could further improve PDT performance under near-infrared (NIR) irradiation. In addition, to their homologous targeting and PA/FL dual-mode imaging ability, MH-PLGA-IR780 NPs had advantages in penetrating deeper into tumor tissues and in real-time dynamic distribution monitoring in vivo, which laid a foundation for further clinical applications in OS. Moreover, we demonstrated that PDT guided by the constructed NPs could significantly induce HOS cells apoptosis and ferroptosis via excessive accumulation of reactive oxygen species (ROS), and further determined that the potential anticancer molecular mechanism of apoptosis was triggered by the release of cytochrome c-activated mitochondrial apoptosis (endogenous apoptosis), and that ferroptosis caused the activation of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and the inactivation of glutathione peroxidase 4 (GPX4), synergistically leading to excessive accumulation of Lipid-ROS and Lipid peroxides (LPOs). Concurrently, MH-PLGA-IR780 NPs-guided PDT also showed an obvious inhibitory effect on tumor growth in vivo. CONCLUSION These results suggest that this homologous targeting-based theranostic nanoplatform provides an effective method to improve PDT performance in OS and contributes a new and promising approach for OS therapy.
Collapse
|
16
|
Niu P, Dai J, Wang Z, Wang Y, Feng D, Li Y, Miao W. Sensitization of Antibiotic-Resistant Gram-Negative Bacteria to Photodynamic Therapy via Perfluorocarbon Nanoemulsion. Pharmaceuticals (Basel) 2022; 15:ph15020156. [PMID: 35215269 PMCID: PMC8878207 DOI: 10.3390/ph15020156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
With the merits of excellent efficacy, safety, and facile implementation, antibacterial photodynamic therapy (APDT) represents a promising means for treating bacterial infections. However, APDT shows an unsatisfactory efficacy in combating antibiotic-resistant Gram-negative bacteria due to their specific cell wall structure. In this work, we report a perfluorocarbon nanoemulsion (Ce6@FDC) used as a multifunctional nanocargo of photosensitizer and oxygen for sensitizing antibiotic-resistant Gram-negative bacteria to APDT. Ce6@FDC was fabricated via ultrasonic emulsification with good colloidal stability, efficient Ce6 and oxygen delivery, and excellent photodynamic activity. Meanwhile, Ce6@FDC could strongly bind with Gram-negative Acinetobacter baumannii (A. baumannii) and Escherichia coli (E. coli) via electrostatic interaction, thus leading to notable photodynamic bactericidal potency upon irradiation. In addition, oxygenated Ce6@FDC also exhibited a remarkable efficacy in eradicating Gram-negative bacteria biofilm, averaging five log units lower than the Ce6 group under identical conditions. Taken together, we demonstrate that photodynamic perfluorocarbon nanoemulsion with oxygen-delivery ability could effectively kill planktonic bacteria and remove biofilm, representing a novel strategy in fighting against antibiotic-resistant Gram-negative bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuanyuan Li
- Correspondence: (Y.L.); (W.M.); Tel.: +86-25-58139399 (W.M.)
| | - Wenjun Miao
- Correspondence: (Y.L.); (W.M.); Tel.: +86-25-58139399 (W.M.)
| |
Collapse
|
17
|
Kong C, Xu B, Qiu G, Wei M, Zhang M, Bao S, Tang J, Li L, Liu J. Multifunctional Nanoparticles-Mediated PTT/PDT Synergistic Immune Activation and Antitumor Activity Combined with Anti-PD-L1 Immunotherapy for Breast Cancer Treatment. Int J Nanomedicine 2022; 17:5391-5411. [PMID: 36419717 PMCID: PMC9677922 DOI: 10.2147/ijn.s373282] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Photoimmunotherapy is a breakthrough treatment for malignant tumors. Its uniqueness is that it uses antibody mediated targeted delivery to achieve high tumor specificity and uses laser-activated biophysical mechanism to accurately induce the rapid death of cancer cells and avoid damaging the surrounding normal tissues. Methods In this paper, an iron-based micelle was designed to encapsulate the photothermal agent indocyanine green (ICG) and a cyclic tripeptide of arginine-glycine-aspartic acid (cRGD) as targeted multifunctional ICG@SANPs-cRGD nanoparticles for combined photothermal/photodynamic/immune therapy of breast cancer. Results The experimental results show that ICG@SANPs-cRGD nanoparticles have good biocompatibility and photothermal conversion ability. Photothermal therapy (PTT) and photodynamic therapy (PDT) based on ICG@SANPs-cRGD can not only inhibit the proliferation, invasion and migration of tumor cells, but also directly kill tumor cells by inducing apoptosis or necrosis. Dual-mode fluorescence light (FL) and magnetic resonance imaging (MRI) imaging in mice confirmed the selective accumulation at tumor sites and imaging ability of ICG@SANPs-cRGD. PTT/PDT combined with Anti-PD-L1 immunotherapy based on ICG@SANPs-cRGD mediated the immunogenic cell death (ICD) of tumor cells by regulating the expression of immune-related indicators and activated the body's immune response mechanism, which enhanced the immunotherapy effect of immune checkpoint block (ICB). PTT/PDT combined with Anti-PD-L1 therapy not only prevented the progression of the primary tumor but also inhibited the distant metastasis of the tumor. Discussion This study explores the biomedical application of PTT/PDT combined with Anti-PD-L1 based on ICG@SANPs-cRGD nanomaterials for breast cancer treatment and demonstrates the potential of ICG@SANPs-cRGD as a multifunctional therapeutic platform for future cancer therapy.
Collapse
Affiliation(s)
- Cunqing Kong
- Department of Ultrasound, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Banghao Xu
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Guanhua Qiu
- Department of Ultrasound, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Meng Wei
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Mengqi Zhang
- Department of Interventional Therapy Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Shengxian Bao
- Department of Ultrasound, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Jiali Tang
- Department of Ultrasound, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - JunJie Liu
- Department of Ultrasound, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| |
Collapse
|
18
|
Zhou J, Hou J, Liu S, Xu J, Luo Y, Zheng J, Li X, Wang Z, Ran H, Guo D. Theranostic Nanoplatform with Sequential SDT and ADV Effects in Response to Well-Programmed LIFU Irradiation for Cervical Cancer. Int J Nanomedicine 2021; 16:7995-8012. [PMID: 34916791 PMCID: PMC8669754 DOI: 10.2147/ijn.s339257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Some patients with cervical cancer have the need to preserve fertility; therefore, a minimally invasive treatment option that can effectively inactivate tumors in these patients is necessary. Methods In this paper, we designed and prepared nanoparticles (NPs) carrying IR780 and perfluorohexane (PFH) and characterized their properties. We focused on the promotion of programmed low-intensity focused ultrasound (LIFU) irradiation on the penetration and treatment of cervical cancer. First we used penetration-enhancing LIFU irradiation to promote the penetration of the NPs into 3D multicellular tumor spheroids (MCTSs) and tumors in tumor-bearing nude mice. Then we used re-therapeutic LIFU irradiation to achieve antitumor effects in vitro and in vivo. Photoacoustic (PA) and magnetic resonance (MR) imaging were used to monitor and evaluate the targeting and therapeutic effects of these NPs on tumor tissues. Results The NPs prepared in this paper exhibited high affinity for HeLa cells, and can selectively achieve mitochondrial localization in the cell due to IR780 assistance. The penetration-enhancing LIFU irradiation have the ability to promote the penetration of the NPs into cervical cancer models in vivo and in vitro. Under LIFU irradiation, the cytotoxic reactive oxygen species (ROS) produced by IR780 during the first half of the re-therapeutic LIFU irradiation and the physical acoustic droplet vaporization (ADV) effect after PFH phase transition during the second half of the re-therapeutic LIFU irradiation can achieve synergistic minimally invasive treatment of tumors, which can be visualized and evaluated by PA and MR imaging in vivo. Conclusion Well-programmed LIFU irradiation can promote NP penetration into deep tumor tissue and achieve antitumor effects simultaneously. Linking ROS + ADV effects can induce cell coagulation necrosis and lead to a comprehensive, long-term impact on tumor tissue, providing a conceptual theranostic nanoplatform for cervical cancer.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jingxin Hou
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.,Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Shuling Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ying Luo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jun Zheng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xin Li
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| |
Collapse
|
19
|
Zhang P, Zhang L, Wang J, Zhu L, Li Z, Chen H, Gao Y. An intelligent hypoxia-relieving chitosan-based nanoplatform for enhanced targeted chemo-sonodynamic combination therapy on lung cancer. Carbohydr Polym 2021; 274:118655. [PMID: 34702474 DOI: 10.1016/j.carbpol.2021.118655] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/23/2021] [Accepted: 09/06/2021] [Indexed: 12/20/2022]
Abstract
The clinical efficacy of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs)-based targeted molecular therapies (TMT) is inevitably hampered by the development of acquired drug resistance in non-small cell lung cancer (NSCLC) treatment. Sonodymanic therapy (SDT) is a promising new cancer treatment approach, but its effects are restricted by tumor hypoxia. Herein, a nanoplatform fabricated by erlotinib-modified chitosan loading sonosensitizer hematoporphyrin (HP) and oxygen-storing agent perfluorooctyl bromide (PFOB), namely CEPH, was developed to deliver HP to erlotinib-sensitive cells. CEPH with ultrasound could alleviate hypoxia inside the three-dimensional multicellular tumor spheroids, suppress NSCLC cell growth under normoxic or hypoxic condition, and enhance TMT/SDT synergistic effects through elevated production of reactive oxygen species, decrease of mitochondrial membrane potential, and down-regulation of the expression of the proteins EGFR, p-EGFR, and HIF-1α. Hence, CEPH could be a potential nanoplatform to improve the efficacy of oxygen-dependent SDT and overcome hypoxia-induced TMT resistance for enhanced synergistic TMT/SDT.
Collapse
Affiliation(s)
- Peixia Zhang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lu Zhang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jun Wang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lisheng Zhu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ziying Li
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Haijun Chen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
20
|
Mena-Giraldo P, Orozco J. Polymeric Micro/Nanocarriers and Motors for Cargo Transport and Phototriggered Delivery. Polymers (Basel) 2021; 13:3920. [PMID: 34833219 PMCID: PMC8621231 DOI: 10.3390/polym13223920] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Smart polymer-based micro/nanoassemblies have emerged as a promising alternative for transporting and delivering a myriad of cargo. Cargo encapsulation into (or linked to) polymeric micro/nanocarrier (PC) strategies may help to conserve cargo activity and functionality when interacting with its surroundings in its journey to the target. PCs for cargo phototriggering allow for excellent spatiotemporal control via irradiation as an external stimulus, thus regulating the delivery kinetics of cargo and potentially increasing its therapeutic effect. Micromotors based on PCs offer an accelerated cargo-medium interaction for biomedical, environmental, and many other applications. This review collects the recent achievements in PC development based on nanomicelles, nanospheres, and nanopolymersomes, among others, with enhanced properties to increase cargo protection and cargo release efficiency triggered by ultraviolet (UV) and near-infrared (NIR) irradiation, including light-stimulated polymeric micromotors for propulsion, cargo transport, biosensing, and photo-thermal therapy. We emphasize the challenges of positioning PCs as drug delivery systems, as well as the outstanding opportunities of light-stimulated polymeric micromotors for practical applications.
Collapse
Affiliation(s)
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 # 52-20, Medellin 050010, Colombia;
| |
Collapse
|
21
|
Wang J, Liu J, Yang Z. Recent advances in peptide-based nanomaterials for targeting hypoxia. NANOSCALE ADVANCES 2021; 3:6027-6039. [PMID: 36133944 PMCID: PMC9418673 DOI: 10.1039/d1na00637a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 06/16/2023]
Abstract
Hypoxia is a prominent feature of many severe diseases such as malignant tumors, ischemic strokes, and rheumatoid arthritis. The lack of oxygen has a paramount impact on angiogenesis, invasion, metastasis, and chemotherapy resistance. The potential of hypoxia as a therapeutic target has been increasingly recognized over the last decade. In order to treat these disease states, peptides have been extensively investigated due to their advantages in safety, target specificity, and tumor penetrability. Peptides can overcome difficulties such as low drug/energy delivery efficiency, hypoxia-induced drug resistance, and tumor nonspecificity. There are three main strategies for targeting hypoxia through peptide-based nanomaterials: (i) using peptide ligands to target cellular environments unique to hypoxic conditions, such as cell surface receptors that are upregulated in cells under hypoxic conditions, (ii) utilizing peptide linkers sensitive to the hypoxic microenvironment that can be cleaved to release therapeutic or diagnostic payloads, and (iii) a combination of the above where targeting peptides will localize the system to a hypoxic environment for it to be selectively cleaved to release its payload, forming a dual-targeting system. This review focuses on recent developments in the design and construction of novel peptide-based hypoxia-targeting nanomaterials, followed by their mechanisms and potential applications in diagnosis and treatment of hypoxic diseases. In addition, we address challenges and prospects of how peptide-based hypoxia-targeting nanomaterials can achieve a wider range of clinical applications.
Collapse
Affiliation(s)
- Jun Wang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Jing Liu
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| | - Zhongxing Yang
- School of Pharmacy, Jining Medical University Rizhao 276800 China
| |
Collapse
|
22
|
Fan X, Ke L, Cheng H, Chen H, Li Z, Ye E, Loh XJ, Wu YL, Liu G, Li Z. Enhanced drug retention by anthracene crosslinked nanocomposites for bimodal imaging-guided phototherapy. NANOSCALE 2021; 13:14713-14722. [PMID: 34473183 DOI: 10.1039/d1nr04171a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Efficient drug delivery, multifunctional combined therapy and real-time diagnosis are the main hallmarks in the exploitation of precision nanomedicine. Herein, an anthracene-functionalized micelle containing a magnetic resonance imaging (MRI) contrast agent, upconversion nanoparticles (UCNPs) and the photosensitizer IR780 is designed to achieve sustained drug release and enhanced photothermal and photodynamic therapy. The polymer-coated hybrid micelle was achieved by crosslinking anthracene-dimer with UV light (λ > 300 nm), which is converted from near-infrared (NIR) irradiation upon UCNPs. Besides, the water-insoluble photosensitizer IR780 is introduced into the system to achieve efficient drug delivery and photothermal and photodynamic synergistic therapy. As a consequence of NIR-induced anthracene-dimer formation, the cross-linked nanocomposite shows sustained drug release, and the enhanced retention effect of IR780 could increase the photothermal conversion efficiency. Importantly, the incorporation of 2,2,6,6-tetramethyl-piperidineoxyl (TEMPO) as a nitroxide MRI contrast agent presents the potential for real-time diagnosis via nanotheranostics, and the fluorescence imaging of IR780 is applied to monitor drug distribution and metabolism. This strategy of sustained drug delivery by anthracene-dimer formation through the better penetration depth of NIR-II fluorescence provides an executable platform to achieve enhanced phototherapy in biomedical applications.
Collapse
Affiliation(s)
- Xiaoshan Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Hu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Enyi Ye
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore 138634, Singapore.
| | - Xian-Jun Loh
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore 138634, Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zibiao Li
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore 138634, Singapore. .,Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, 117576, Singapore
| |
Collapse
|
23
|
Wang X, Li Y, Deng X, Jia F, Cui X, Lu J, Pan Z, Wu Y. Colloidally Stabilized DSPE-PEG-Glucose/Calcium Phosphate Hybrid Nanocomposites for Enhanced Photodynamic Cancer Therapy via Complementary Mitochondrial Ca 2+ Overload and Autophagy Inhibition. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39112-39125. [PMID: 34384220 DOI: 10.1021/acsami.1c11583] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Autophagy inhibition could hinder the underlying protective mechanisms in the course of tumor treatment. The advances in autophagy inhibition have driven focus on the functionalized nanoplatforms by combining the current treatment paradigms with complementary autophagy inhibition for enhanced efficacy. Furthermore, Ca2+ overload is also a promising adjuvant target for the tumor treatment by augmenting mitochondrial damage. In this view, complementary mitochondrial Ca2+ overload and autophagy inhibition were first demonstrated as a novel strategy suitable for homing in on the shortage of photodynamic therapy (PDT). We constructed biodegradable tumor-targeted inorganic/organic hybrid nanocomposites (DPGC/OI) synchronously encapsulating IR780 and Obatoclax by biomineralization of the nanofilm method, which consists of pH-triggered calcium phosphate (CP), long circulation phospholipid block copolymers 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-poly(ethylene glycol) (PEG)2000-glucose (DPG). In the presence of the hydrophilic PEG chain and glucose transporter 1 (Glut-1) ligands, DPGC would become an effectively tumor-oriented nanoplatform. Subsequently, IR780 as an outstanding photosensitizer could produce increased amounts of toxic reactive oxygen species (ROS) after laser irradiation. Calcium phosphate (CP) as the Ca2+ nanogenerator could generate Ca2+ at low pH to induce mitochondrial Ca2+ overload. The dysfunction of mitochondria could enhance increased amounts of ROS. Based on the premise that autophagy would degrade dysfunctional organelles to sustain metabolism and homeostasis, which might participate in resistance to PDT, Obatoclax as an autophagy inhibitor would hinder the protective mechanism from cancer cells with negligible toxicity. Such an enhanced PDT via mitochondrial Ca2+ overload and autophagy inhibition could be realized by DPGC/OI.
Collapse
Affiliation(s)
- Xuan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yunhao Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Xiongwei Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
| | - Fan Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xinyue Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
| | - Jianqing Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
| | - Zian Pan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
24
|
Zheng Y, Ye J, Li Z, Chen H, Gao Y. Recent progress in sono-photodynamic cancer therapy: From developed new sensitizers to nanotechnology-based efficacy-enhancing strategies. Acta Pharm Sin B 2021; 11:2197-2219. [PMID: 34522584 PMCID: PMC8424231 DOI: 10.1016/j.apsb.2020.12.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/27/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022] Open
Abstract
Many sensitizers have not only photodynamic effects, but also sonodynamic effects. Therefore, the combination of sonodynamic therapy (SDT) and photodynamic therapy (PDT) using sensitizers for sono-photodynamic therapy (SPDT) provides alternative opportunities for clinical cancer therapy. Although significant advances have been made in synthesizing new sensitizers for SPDT, few of them are successfully applied in clinical settings. The anti-tumor effects of the sensitizers are restricted by the lack of tumor-targeting specificity, incapability in deep intratumoral delivery, and the deteriorating tumor microenvironment. The application of nanotechnology-based drug delivery systems (NDDSs) can solve the above shortcomings, thereby improving the SPDT efficacy. This review summarizes various sensitizers as sono/photosensitizers that can be further used in SPDT, and describes different strategies for enhancing tumor treatment by NDDSs, such as overcoming biological barriers, improving tumor-targeted delivery and intratumoral delivery, providing stimuli-responsive controlled-release characteristics, stimulating anti-tumor immunity, increasing oxygen supply, employing different therapeutic modalities, and combining diagnosis and treatment. The challenges and prospects for further development of intelligent sensitizers and translational NDDSs for SPDT are also discussed.
Collapse
Affiliation(s)
- Yilin Zheng
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jinxiang Ye
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350116, China
| | - Ziying Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Haijun Chen
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350116, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fuzhou University, Fuzhou 350116, China
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
25
|
Lu F, Li Z, Sheng Y, Ma Y, Yang Y, Ren Y, Su Z, Yu R, Zhang S. Thermal-triggered packing of lipophilic NIR dye IR780 in hepatitis B core at critical ionic strength and cargo-host ratio for improved stability and enhanced cancer phototherapy. Biomaterials 2021; 276:121035. [PMID: 34303153 DOI: 10.1016/j.biomaterials.2021.121035] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 02/09/2023]
Abstract
Virus-like particles (VLPs) holding internal cavity with diameter from tens up to one hundred nanometers are attractive platform for drug delivery. Nevertheless, the packing of drugs in the nanocage mainly relies on complicated disassembly-reassembly process. In this study, hepatitis B core protein (HBc) VLPs which can withstand temperature up to 90 °C was employed as carrier to load a lipophilic near infrared dye IR780. It was found that an attaching-dis-atching-diffusing process was involved for the entering of IR780 in the cavity of HBc. The first two steps were associated with the electrostatic interactions between oppositely charged HBc and IR780, which was critically manipulated by ionic strength and HBc/IR780 mass ratio at which they were mixed; while the diffusion of IR780 across the shell of HBc showed a temperature-dependent manner that can be triggered by thermal induced pore-opening of the HBc capsid. At optimized condition, about 1055 IR780 molecules were encapsulated in each HBc by simply mixing them for 10 min at 60 °C. Compared with free IR780, the HBc-IR780 particles showed significantly improved aqueous and photostability, as well as enhanced photothermal and photodynamic performance for cancer therapy. This study provides a novel drug loading strategy and nanomemedicine for cancer phototherapies.
Collapse
Affiliation(s)
- Fengying Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhengjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yanan Sheng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yanyan Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yanli Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ying Ren
- State Key Laboratory of Multiphase Complex System, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhiguo Su
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Rong Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
26
|
Zhou L, Pi W, Hao M, Li Y, An H, Li Q, Zhang P, Wen Y. An injectable and biodegradable nano-photothermal DNA hydrogel enhances penetration and efficacy of tumor therapy. Biomater Sci 2021; 9:4904-4921. [PMID: 34047319 DOI: 10.1039/d1bm00568e] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biological barrier of solid tumors hinders deep penetration of nanomedicine, constraining anticancer treatment. Moreover, the inherent multidrug resistance (MDR) of cancer tissues may further limit the efficacy of anti-tumor nanomedicine. We synthesized highly permeable, photothermal, injectable, and positively charged biodegradable nucleic acid hydrogel (DNA-gel) nanoparticles to deliver cancer drugs. The nanoparticles are derived from photothermal materials containing black phosphorus quantum dots (BPQDs). The intra-tumoral BPQDs improve the sensitivity of tumor cells to photothermal therapy (PTT) and photodynamic treatment (PDT). Tumor cells take up the positively charged and controllable size DNA-gel nanoparticles, facilitating easy penetration and translocation of the particles across and within the cells. Mouse models demonstrated the anti-tumor activity of the DNA gel nanoparticles in vivo. In particular, the DNA gel nanoparticles enhanced clearance of both small and large tumor masses. Just 20 days after treatment, the tumor masses had been cleared. Compared to DOX chemotherapy alone, the DNA-gel treatment also significantly reduced drug resistance and improved the overall survival of mice with orthotopic breast tumors (83.3%, 78 d). Therefore, DNA gel nanoparticles are safe and efficient supplements for cancer therapy.
Collapse
Affiliation(s)
- Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Wei Pi
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Mingda Hao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Yansheng Li
- Beijing Key Laboratory for Sensors, Beijing Information Science & Technology University, Beijing 100192, China
| | - Heng An
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Qicheng Li
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Peixun Zhang
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| |
Collapse
|
27
|
Abstract
IR780, a small molecule with a strong optical property and excellent photoconversion efficiency following near infrared (NIR) irradiation, has attracted increasing attention in the field of cancer treatment and imaging. This review is focused on different IR780-based nanoplatforms and the application of IR780-based nanomaterials for cancer bioimaging and therapy. Thus, this review summarizes the overall aspects of IR780-based nanomaterials that positively impact cancer biomedical applications.
Collapse
Affiliation(s)
- Long Wang
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China. and Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chengcheng Niu
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China. and Department of Ultrasound Diagnosis and Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
28
|
Liu J, Zhao X, Nie W, Yang Y, Wu C, Liu W, Zhang K, Zhang Z, Shi J. Tumor cell-activated "Sustainable ROS Generator" with homogeneous intratumoral distribution property for improved anti-tumor therapy. Am J Cancer Res 2021; 11:379-396. [PMID: 33391481 PMCID: PMC7681092 DOI: 10.7150/thno.50028] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Photodynamic therapy (PDT) holds a number of advantages for tumor therapy. However, its therapeutic efficiency is limited by non-sustainable reactive oxygen species (ROS) generation and heterogeneous distribution of photosensitizer (PS) in tumor. Herein, a "Sustainable ROS Generator" (SRG) is developed for efficient antitumor therapy. Methods: SRG was prepared by encapsulating small-sized Mn3O4-Ce6 nanoparticles (MC) into dendritic mesoporous silica nanoparticles (DMSNs) and then enveloped with hyaluronic acid (HA). Due to the high concentration of HAase in tumor tissue, the small-sized MC could be released from DMSNs and homogeneously distributed in whole tumor. Then, the released MC would be uptaken by tumor cells and degraded by high levels of intracellular glutathione (GSH), disrupting intracellular redox homeostasis. More importantly, the released Ce6 could efficiently generate singlet oxygen (1O2) under laser irradiation until the tissue oxygen was exhausted, and the manganese ion (Mn2+) generated by degraded MC would then convert the low toxic by-product (H2O2) of PDT to the most harmful ROS (·OH) for sustainable and recyclable ROS generation. Results: MC could be homogeneously distributed in whole tumor and significantly reduced the level of intracellular GSH. At 2 h after PDT, obvious intracellular ROS production was still observed. Moreover, during oxygen recovery in tumor tissue, ·OH could be continuously produced, and the nanosystem could induce 82% of cell death comparing with 30% of cell death induced by free Ce6. For in vivo PDT, SRG achieved a complete inhibition on tumor growth. Conclusion: Based on these findings, we conclude that the designed SRG could induce sustainable ROS generation, homogeneous intratumoral distribution and intracellular redox homeostasis disruption, presenting an efficient strategy for enhanced ROS-mediated anti-tumor therapy.
Collapse
|
29
|
Wang Y, Luo S, Wu Y, Tang P, Liu J, Liu Z, Shen S, Ren H, Wu D. Highly Penetrable and On-Demand Oxygen Release with Tumor Activity Composite Nanosystem for Photothermal/Photodynamic Synergetic Therapy. ACS NANO 2020; 14:17046-17062. [PMID: 33290657 DOI: 10.1021/acsnano.0c06415] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A deep penetrating and pH-responsive composite nanosystem was strategically developed to improve the efficacy of synergetic photothermal/photodynamic therapy (PTT/PDT) against hypoxic tumor. The designed nanosystem ([PHC]PP@HA NPs) was constructed by coloading hemoglobin (Hb) and chlorin e6 on polydopamine to build small-sized PHC NPs, which were encapsulated inside the polymer micelles (poly(ethylene glycol)-poly(ethylenimine)) and then capped with functionalized hyaluronic acid. The pH-responsive feature made [PHC]PP@HA NPs retain an initial size of ∼140 nm in blood circulation but rapidly release small PHC NPs (∼10 nm) with a high tumor-penetrating ability in the tumor microenvironment. The in vitro penetration experiment showed that the penetration depth of PHC NPs in the multicellular tumor spheroids exceeded 110 μm. The [PHC]PP@HA NPs exhibited excellent biocompatibility, deep tumor permeability, high photothermal conversion efficiency (47.09%), and low combination index (0.59) under hypoxic conditions. Notably, the nanosystem can freely adjust the release of oxygen and damaging PHC NPs in an on-demand manner on the basis of the feedback of tumor activity. This feedback tumor therapy significantly improved the synergistic effect of PTT/PDT and reduced its toxic side effects. The in vivo antitumor results showed that the tumor inhibition rate of [PHC]PP@HA NPs with an on-demand oxygen supply of Hb was ∼100%, which was much better than those of PTT alone and Hb-free nanoparticles ([PC]PP@HA NPs). Consequently, the [PHC]PP@HA NP-mediated PTT/PDT guided by feedback tumor therapy achieved an efficient tumor ablation with an extremely low tumor recurrence rate (8.3%) 60 d later, indicating the versatile potential of PTT/PDT.
Collapse
Affiliation(s)
- Ya Wang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Siyuan Luo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Youshen Wu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Peng Tang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Jiajun Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zeying Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Shihong Shen
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Haozhe Ren
- Health Science Center, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P. R. China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
30
|
Recent advances of sorafenib nanoformulations for cancer therapy: Smart nanosystem and combination therapy. Asian J Pharm Sci 2020; 16:318-336. [PMID: 34276821 PMCID: PMC8261086 DOI: 10.1016/j.ajps.2020.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/01/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022] Open
Abstract
Sorafenib, a molecular targeted multi-kinase inhibitor, has received considerable interests in recent years due to its significant profiles of efficacy in cancer therapy. However, poor pharmacokinetic properties such as limited water solubility, rapid elimination and metabolism lead to low bioavailability, restricting its further clinical application. Over the past decade, with substantial progress achieved in the development of nanotechnology, various types of smart sorafenib nanoformulations have been developed to improve the targetability as well as the bioavailability of sorafenib. In this review, we summarize various aspects from the preparation and characterization to the evaluation of antitumor efficacy of numerous stimuli-responsive sorafenib nanodelivery systems, particularly with emphasis on their mechanism of drug release and tumor microenvironment response. In addition, this review makes great effort to summarize the nanosystem-based combination therapy of sorafenib with other antitumor agents, which can provide detailed information for further synergistic cancer therapy. In the final section of this review, we also provide a detailed discussion of future challenges and prospects of designing and developing ideal sorafenib nanoformulations for clinical cancer therapy.
Collapse
|
31
|
Wang X, Ding X, Yu B, Zhang X, Shen Y, Cong H. Tumor microenvironment-responsive polymer with chlorin e6 to interface hollow mesoporous silica nanoparticles-loaded oxygen supply factor for boosted photodynamic therapy. NANOTECHNOLOGY 2020; 31:305709. [PMID: 32299065 DOI: 10.1088/1361-6528/ab89d1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer treatment has always been a big problem for people. With the application of photodynamic therapy, the problem has been alleviated. However, the problem of tumor hypoxia affecting photodynamic therapy has been waiting to be resolved. Therefore, we report here that a redox nanocarrier (called RN) is prepared by hollow mesoporous silica sphere (HMSNs) and a redox-responsive polymer ligand. The nanocarrier is loaded with metformin and catalase, and the polymer is linked to the photosensitizer chlorin e6 (Ce6). Metformin inhibits the mitochondrial respiration of cancer cells, reducing the activity of cancer cells and increasing the oxygen concentration required for photodynamic therapy. Not only the effect of photodynamic therapy is enhanced, but also the effect of chemotherapy is increased to achieve super additive treatment. These RNs exhibit not only low cytotoxicity but also high biocompatibility in vitro experiments. In vitro Ce6 release studies have shown a higher release in the presence of glutathione (GSH). Confocal microscopy can further indicate that the nanoparticles are carried to the area around the nucleus of the cancer cells. In addition, treatment with a mouse tumor model demonstrated that RN has an effective therapeutic effect on tumors.
Collapse
Affiliation(s)
- Xuemei Wang
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, People's Republic of China
| | | | | | | | | | | |
Collapse
|
32
|
Ge X, Fu Q, Su L, Li Z, Zhang W, Chen T, Yang H, Song J. Light-activated gold nanorod vesicles with NIR-II fluorescence and photoacoustic imaging performances for cancer theranostics. Theranostics 2020; 10:4809-4821. [PMID: 32308751 PMCID: PMC7163452 DOI: 10.7150/thno.44376] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
Fluorescence (FL) and photoacoustic (PA) imaging in the second near infrared window (NIR-II FL and NIR-II PA) hold great promise for biomedical applications because of their non-invasive nature and excellent spatial resolution properties. Methods: We develop a NIR-II PA and NIR-II FL dual-mode imaging gold nanorod vesicles (AuNR Ves) by self-assembly of amphiphilic AuNR coated with light responsive polyprodrug of Ru-complex and PEG, and NIR-II cyanine dye (IR 1061). The AuNR Ves showed strong ligh absorption property and PA imaging performance in the NIR-II windows. Moreover, the NIR-II fluorescence signal of IR 1061 loaded in the AuNR Ve is quenched. Results: The AuNR Ves can release photosensitizer Ru-complex and IR 1061 sequentially triggered by NIR light irradiation, leading to a corresponding NIR-II PA signal decrease and NIR-II FL signal recovery. Meanwhile, Ru-complex can not only serve as a chemotherapeutic drug but also generate singlet oxygen (1O2) under NIR light irradiation. The release of Ru-complex and photodynamic therapy are guided by the responsive variation of NIR-II PA and NIR-II FL signals. Conclusions: The AuNR Ve possessing not only precisely control 1O2/drug release but also the intrinsic ability to monitor therapy process offers a new strategy for the development of smart theranostic nanoplatform.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jibin Song
- MOE key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
33
|
Leitão MM, de Melo‐Diogo D, Alves CG, Lima‐Sousa R, Correia IJ. Prototypic Heptamethine Cyanine Incorporating Nanomaterials for Cancer Phototheragnostic. Adv Healthc Mater 2020; 9:e1901665. [PMID: 31994354 DOI: 10.1002/adhm.201901665] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Developing technologies that allow the simultaneous diagnosis and treatment of cancer (theragnostic) has been the quest of numerous interdisciplinary research teams. In this context, nanomaterials incorporating prototypic near infrared (NIR)-light responsive heptamethine cyanines have been showing very promising results for cancer theragnostic. The precisely engineered features of these nanomaterials endow them with the ability to achieve a high tumor accumulation, enabling a tumor's visualization by NIR fluorescence and photoacoustic imaging modalities. Upon interaction with NIR light, the tumor-homed heptamethine cyanine-incorporating nanomaterials can also produce a photothermal/photodynamic effect with a high spatio-temporal resolution and minimal side effects, leading to an improved therapeutic outcome. This progress report analyses the application of nanomaterials incorporating prototypic NIR-light responsive heptamethine cyanines (IR775, IR780, IR783, IR797, IR806, IR808, IR820, IR825, IRDye 800CW, and Cypate) for cancer photothermal therapy, photodynamic therapy, and imaging. Overall, the continuous development of nanomaterials incorporating the prototypic NIR absorbing heptamethine cyanines will cement their phototheragnostic capabilities.
Collapse
Affiliation(s)
- Miguel M. Leitão
- CICS‐UBI‐Centro de Investigação em Ciências da SaúdeUniversidade da Beira Interior 6200‐506 Covilhã Portugal
| | - Duarte de Melo‐Diogo
- CICS‐UBI‐Centro de Investigação em Ciências da SaúdeUniversidade da Beira Interior 6200‐506 Covilhã Portugal
| | - Cátia G. Alves
- CICS‐UBI‐Centro de Investigação em Ciências da SaúdeUniversidade da Beira Interior 6200‐506 Covilhã Portugal
| | - Rita Lima‐Sousa
- CICS‐UBI‐Centro de Investigação em Ciências da SaúdeUniversidade da Beira Interior 6200‐506 Covilhã Portugal
| | - Ilídio J. Correia
- CICS‐UBI‐Centro de Investigação em Ciências da SaúdeUniversidade da Beira Interior 6200‐506 Covilhã Portugal
- CIEPQPF‐Departamento de Engenharia QuímicaUniversidade de CoimbraRua Sílvio Lima 3030‐790 Coimbra Portugal
| |
Collapse
|
34
|
Hu D, Pan M, Yu Y, Sun A, Shi K, Qu Y, Qian Z. Application of nanotechnology for enhancing photodynamic therapy via ameliorating, neglecting, or exploiting tumor hypoxia. VIEW 2020. [DOI: 10.1002/viw2.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- DanRong Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - Meng Pan
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - Yan Yu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - Ao Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - Ying Qu
- Department of Hematology and Research Laboratory of HematologyState Key Laboratory of BiotherapyWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University, Collaborative Innovation Center for Biotherapy Chengdu Sichuan P. R. China
| |
Collapse
|
35
|
Zhao L, Fu C, Tan L, Li T, Zhong H, Meng X. Advanced nanotechnology for hypoxia-associated antitumor therapy. NANOSCALE 2020; 12:2855-2874. [PMID: 31965135 DOI: 10.1039/c9nr09071a] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Hypoxia is a hallmark of the tumor microenvironment, which promotes the proliferation, metastasis and invasion of tumors and stimulates the resistance of cancer treatments, leading to the serious consequence of tumor recurrence. Many nanotechnology-based studies have been conducted to improve the efficacy of cancer treatments using a hypoxia strategy. This is usually achieved by (i) activating bioreductive prodrugs in the tumor hypoxic/exacerbated hypoxic microenvironment, or (ii) delivering therapeutic agents to hypoxic tumor tissue using targeting molecules. Normally, a good therapeutic effect can be expected upon modulating the hypoxic microenvironment for tumor treatments. To achieve this, various nanotechnology strategies based on overcoming hypoxia have been exploited to alleviate tumor hypoxia and enhance the therapeutic efficacy of tumor therapy, including (i) reducing oxygen consumption by inhibiting cell respiration, (ii) normalizing tumor vessels to promote blood flow in the tumor, (iii) carrying exogenous oxygen into the tumor, and (iv) generating oxygen in situ. The strategy of in situ oxygen production is refined, and the scope of this strategy is further expanded. Finally, the inspiration of using advanced nanotechnology in hypoxia-associated antitumor therapy guides the study of tumor hypoxia for clinical use.
Collapse
Affiliation(s)
- Lirong Zhao
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Number 29 East Road Zhongguancun, Beijing 100190, P. R. China. and University of the Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Changhui Fu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Number 29 East Road Zhongguancun, Beijing 100190, P. R. China.
| | - Longfei Tan
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Number 29 East Road Zhongguancun, Beijing 100190, P. R. China.
| | - Ting Li
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Number 29 East Road Zhongguancun, Beijing 100190, P. R. China. and University of the Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongshan Zhong
- Department of Radiology, First Hospital of China Medical University, Shenyang 110001, P. R. China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Number 29 East Road Zhongguancun, Beijing 100190, P. R. China.
| |
Collapse
|
36
|
Wang J, Zhang B, Sun J, Wang Y, Wang H. Nanomedicine-Enabled Modulation of Tumor Hypoxic Microenvironment for Enhanced Cancer Therapy. ADVANCED THERAPEUTICS 2020; 3:1900083. [PMID: 34277929 PMCID: PMC8281934 DOI: 10.1002/adtp.201900083] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Indexed: 01/21/2023]
Abstract
Hypoxia is a common condition of solid tumors that is mainly caused by enhanced tumor proliferative activity and dysfunctional vasculature. In the treatment of hypoxic human solid tumors, many conventional therapeutic approaches (e.g., oxygen-dependent photodynamic therapy, anticancer drug-based chemotherapy or X-ray induced radiotherapy) become considerably less effective or ineffective. In recent years, various strategies have been explored to deliver or generate oxygen inside solid tumors to overcome tumorous hypoxia and show promising evidence to improve the antitumor efficiency. In this review, the extrinsic regulation of tumor hypoxia via nanomaterial delivery is discussed followed by a summary of the mechanisms through which the modulated tumor hypoxic microenvironment improves therapeutic efficacy. The review concludes with future perspectives, to specifically address the translation of nanomaterial-based therapeutic strategies for clinical applications.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA; Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
37
|
Wang H, Li J, Wang Y, Gong X, Xu X, Wang J, Li Y, Sha X, Zhang Z. Nanoparticles-mediated reoxygenation strategy relieves tumor hypoxia for enhanced cancer therapy. J Control Release 2019; 319:25-45. [PMID: 31862359 DOI: 10.1016/j.jconrel.2019.12.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Abstract
Tumor hypoxia is a characteristic hallmark of malignant solid tumors, which remains an essential impediment to many current treatments like chemotherapy, radiotherapy, photodynamic therapy and immunotherapy, thereby leading to poor clinical prognosis after therapy. Rationally, modulating tumor hypoxia can be of great interest to augment the therapeutic efficacy of these treatments. In this review, we focus our discussion on current advances in nanoparticles-mediated tumor reoxygenation strategy for relieving tumor hypoxia to improve the therapeutic efficacy of versatile therapies. These nanoparticles can improve tumor oxygen levels via nanoparticles-mediated oxygen-carrying or oxygen-generating tactics to synergize the effectiveness of many current therapeutic modalities. Based on these considerable summaries and analyses, we propose some feasible perspectives on nanoparticles-based tumor reoxygenations to ameliorate the therapeutic outcomes.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Yantai University, Shandong 264000, China.
| | - Xianyi Sha
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Shandong 264000, China.
| |
Collapse
|
38
|
Wu H, Wang C, Sun J, Sun L, Wan J, Wang S, Gu D, Yu C, Yang C, He J, Zhang Z, Lv Y, Wang H, Yao M, Qin W, Wang C, Jin H. Self-Assembled and Self-Monitored Sorafenib/Indocyanine Green Nanodrug with Synergistic Antitumor Activity Mediated by Hyperthermia and Reactive Oxygen Species-Induced Apoptosis. ACS APPLIED MATERIALS & INTERFACES 2019; 11:43996-44006. [PMID: 31682099 DOI: 10.1021/acsami.9b18086] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liver cancer is a leading cause of cancer morbidity and mortality worldwide, especially in China. Sorafenib (SRF) is currently the most commonly used systemic agent against advanced hepatocellular carcinoma (HCC), which is the most common type of liver cancer. However, HCC patients have only limited benefit and suffer a serious side effect from SRF. Therefore, new approaches are urgently needed to improve the therapeutic effectiveness of SRF and reduce its side effect. In our current study, we developed a self-imaging and self-delivered nanodrug with SRF and indocyanine (ICG) to improve the therapeutic effect of sorafenib against HCC. With the π-π stacking effect between SRF and ICG, a one-step nanoprecipitation method was designed to obtain the SRF/ICG nanoparticles (SINP) via self-assembly. Pluronic F127 was used to shield the SINP to further improve the stability in an aqueous environment. The stability, photothermal effect, cell uptake, ROS production, cytotoxicity, tumor imaging, and tumor-targeting and tumor-killing efficacy of the SINP were evaluated in vitro and in vivo by using an HCC cell line Huh7 and its xenograft tumor model. We found that our designed SINP showed monodisperse stability and efficient photothermal effect both in vitro and in vivo. SINP could rapidly enter Huh7 cells and achieve potent cytotoxicity under near-infrared (NIR) laser irradiation partly by producing a great amount of reactive oxygen species (ROS). SINP had significantly improved stability and blood half-life, and could specifically target tumor via the enhanced permeability and retention (EPR) effect in vivo. In addition, SINP showed improved cytotoxicity in both subcutaneous and orthotopic HCC implantation models in vivo. Overall, this rationally designed sorafenib delivery system with a very high loading capacity (33%) has considerably improved antitumor efficiency in vitro and could completely eliminate subcutaneous tumors without any regrowth in vivo. In conclusion, our self-imaging and self-delivered nanodrug could improve the efficacy of SRF and might be a potential therapy for HCC patients.
Collapse
Affiliation(s)
- Haiqiu Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Jiaxin Sun
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Luyan Sun
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Jiaxun Wan
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Siying Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Dishui Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
- Department of Pathophysiology, School of Basic Medical Sciences , Guangdong Medical University , Dongguan , Guangdong 523808 , China
| | - Chengtao Yu
- School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai 200032 , China
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
- Shanghai Medical College , Fudan University , Shanghai 200032 , China
| | - Jia He
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Zihao Zhang
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Yuanyuan Lv
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| | - Changchun Wang
- State Key Laboratory of Molecular Engineering of Polymers, and Department of Macromolecular Science, Laboratory of Advanced Materials , Fudan University , Shanghai 200433 , China
| | - Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute , Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200032 , China
| |
Collapse
|
39
|
Larue L, Myrzakhmetov B, Ben-Mihoub A, Moussaron A, Thomas N, Arnoux P, Baros F, Vanderesse R, Acherar S, Frochot C. Fighting Hypoxia to Improve PDT. Pharmaceuticals (Basel) 2019; 12:E163. [PMID: 31671658 PMCID: PMC6958374 DOI: 10.3390/ph12040163] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 10/24/2019] [Accepted: 10/26/2019] [Indexed: 12/11/2022] Open
Abstract
Photodynamic therapy (PDT) has drawn great interest in recent years mainly due to its low side effects and few drug resistances. Nevertheless, one of the issues of PDT is the need for oxygen to induce a photodynamic effect. Tumours often have low oxygen concentrations, related to the abnormal structure of the microvessels leading to an ineffective blood distribution. Moreover, PDT consumes O2. In order to improve the oxygenation of tumour or decrease hypoxia, different strategies are developed and are described in this review: 1) The use of O2 vehicle; 2) the modification of the tumour microenvironment (TME); 3) combining other therapies with PDT; 4) hypoxia-independent PDT; 5) hypoxia-dependent PDT and 6) fractional PDT.
Collapse
Affiliation(s)
- Ludivine Larue
- Laboratoire Réactions et Génie des Procédés (LRGP), UMR 7274, CNRS, Université de Lorraine, 54000 Nancy, France.
| | | | - Amina Ben-Mihoub
- Laboratoire de Chimie Physique Macromoléculaire (LCPM), UMR 7375, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Albert Moussaron
- Laboratoire Réactions et Génie des Procédés (LRGP), UMR 7274, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Noémie Thomas
- Biologie, Signaux et Systèmes en Cancérologie et Neurosciences, CRAN, UMR 7039, Université de Lorraine, CNRS, 54000 Nancy, France.
| | - Philippe Arnoux
- Laboratoire Réactions et Génie des Procédés (LRGP), UMR 7274, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Francis Baros
- Laboratoire Réactions et Génie des Procédés (LRGP), UMR 7274, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Régis Vanderesse
- Laboratoire de Chimie Physique Macromoléculaire (LCPM), UMR 7375, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Samir Acherar
- Laboratoire de Chimie Physique Macromoléculaire (LCPM), UMR 7375, CNRS, Université de Lorraine, 54000 Nancy, France.
| | - Céline Frochot
- Laboratoire Réactions et Génie des Procédés (LRGP), UMR 7274, CNRS, Université de Lorraine, 54000 Nancy, France.
| |
Collapse
|
40
|
Yang G, Tian J, Chen C, Jiang D, Xue Y, Wang C, Gao Y, Zhang W. An oxygen self-sufficient NIR-responsive nanosystem for enhanced PDT and chemotherapy against hypoxic tumors. Chem Sci 2019; 10:5766-5772. [PMID: 31293763 PMCID: PMC6568044 DOI: 10.1039/c9sc00985j] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/22/2019] [Indexed: 12/30/2022] Open
Abstract
The efficacy of photodynamic therapy and chemotherapy is largely limited by oxygen deficiency in the hypoxic tumor microenvironment. To solve these problems, we fabricated a novel NIR-responsive nanosystem which could co-deliver oxygen and anticancer drug DOX. An oxygen self-sufficient amphiphile (F-IR780-PEG) was first synthesized and subsequently utilized to load anticancer drug DOX to form nanoparticles (F/DOX nanoparticles). Due to the high oxygen capacity of such nanoparticles, the hypoxic tumor microenvironment was greatly modulated after these nanoparticles reached the tumor region, and the results revealed that hypoxia-inducible factor α (HIF-1α) was down-regulated and the expression of P-glycoprotein (P-gp) was then reduced, which were in favor of chemotherapy. Under light irradiation at 808 nm, IR780 could efficiently produce singlet oxygen to damage cancer cells by photodynamic therapy (PDT). Simultaneously, the IR780 linkage could be cleaved by singlet oxygen generated by itself and resulted in DOX release, which further caused cell damage by chemotherapy. With the combination of PDT and chemotherapy, F/DOX nanoparticles showed remarkable therapeutic efficacy under in vitro and in vivo conditions. Furthermore, the F/DOX nanoparticles are favorable for imaging-guided tumor therapy due to the inherent fluorescence properties of IR780. We thus believe that the synergistic treatment described here leads to an ideal therapeutic approach to hypoxic tumors.
Collapse
Affiliation(s)
- Guoliang Yang
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Chao Chen
- State Key Laboratory of Bioreactor Engineering Center , East China University of Science and Technology , China
| | - Dawei Jiang
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Yudong Xue
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Chaochao Wang
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Yun Gao
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry , Key Laboratory for Specially Functional Polymeric Materials and Related Technology of the Ministry of Education , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , China .
| |
Collapse
|
41
|
Liu X, Tian K, Zhang J, Zhao M, Liu S, Zhao Q, Huang W. Smart NIR-Light-Mediated Nanotherapeutic Agents for Enhancing Tumor Accumulation and Overcoming Hypoxia in Synergistic Cancer Therapy. ACS APPLIED BIO MATERIALS 2019; 2:1225-1232. [DOI: 10.1021/acsabm.8b00790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xiangmei Liu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Kang Tian
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Jinghui Zhang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Menglong Zhao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Shujuan Liu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Qiang Zhao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing 210023, P. R. China
| | - Wei Huang
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi’an 710072, Shaanxi China
| |
Collapse
|
42
|
Functional Nanoparticles for Tumor Penetration of Therapeutics. Pharmaceutics 2018; 10:pharmaceutics10040193. [PMID: 30340364 PMCID: PMC6321075 DOI: 10.3390/pharmaceutics10040193] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/16/2022] Open
Abstract
Theranostic nanoparticles recently received great interest for uniting unique functions to amplify therapeutic efficacy and reduce side effects. Despite the enhanced permeability and retention (EPR) effect, which amplifies the accumulation of nanoparticles at the site of a tumor, tumor heterogeneity caused by the dense extracellular matrix of growing cancer cells and the interstitial fluid pressure from abnormal angiogenesis in the tumor inhibit drug/particle penetration, leading to inhomogeneous and limited treatments. Therefore, nanoparticles for penetrated delivery should be designed with different strategies to enhance efficacy. Many strategies were developed to overcome the obstacles in cancer therapy, and they can be divided into three main parts: size changeability, ligand functionalization, and modulation of the tumor microenvironment. This review summarizes the results of ameliorated tumor penetration approaches and amplified therapeutic efficacy in nanomedicines. As the references reveal, further study needs to be conducted with comprehensive strategies with broad applicability and potential translational development.
Collapse
|