151
|
Xu C, Xu L, Han R, Zhu Y, Zhang J. Blood circulation stable doxorubicin prodrug nanoparticles containing hydrazone and thioketal moieties for antitumor chemotherapy. Colloids Surf B Biointerfaces 2021; 201:111632. [PMID: 33667865 DOI: 10.1016/j.colsurfb.2021.111632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/26/2021] [Accepted: 02/15/2021] [Indexed: 12/21/2022]
Abstract
Prodrug nanoparticles with cleavable moieties sensitive to intracellular stimuli have drawn great attention on cancer chemotherapy. Herein, a reactive oxygen species (ROS)-responsive doxorubicin prodrug mPEG-Phe-TK-Phe-hyd-DOX was synthesized, in which hydrophilic methoxy poly(ethylene glycol) (mPEG) and hydrophobic anticancer drug doxorubicin (DOX) were conjugated with hydrazone (hyd) and ROS-responsive thioketal (TK) moieties. The ROS-responsiveness of prodrug was confirmed by proton nuclear magnetic resonance (1H NMR) and dynamic light scattering (DLS). Unexpectedly, the results of in vitro drug release indicated that the hydrazone bond of prodrug nanoparticles was insensitive to pH, which may be due to the strong hydrophobicity, π-π interactions and cation-π interactions jointly inhibited the hydrolysis of hydrazone bonds under acidic conditions. The cellular uptake and in vitro anticancer study showed that ROS-responsive prodrug nanoparticles exhibited faster cellular uptake and better anticancer efficacy. The in vivo experiments showed that the ROS-responsive prodrug nanoparticles had comparable antitumor efficacy with free anticancer drug DOX and reduced organ toxicity. Our results provide novel idea of successfully design multi-stimuli-responsive nano-drug carrier.
Collapse
Affiliation(s)
- Caidie Xu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Long Xu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China.
| | - Renlu Han
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China
| | - Yabin Zhu
- Medical School of Ningbo University, Ningbo, 315211, China
| | - Jianfeng Zhang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
152
|
Lin X, Huang R, Huang Y, Wang K, Li H, Bao Y, Wu C, Zhang Y, Tian X, Wang X. Nanosonosensitizer-Augmented Sonodynamic Therapy Combined with Checkpoint Blockade for Cancer Immunotherapy. Int J Nanomedicine 2021; 16:1889-1899. [PMID: 33707944 PMCID: PMC7943542 DOI: 10.2147/ijn.s290796] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Sonodynamic therapy (SDT) has good targeting and non-invasive advantages in the treatment of solid cancers, and checkpoint blockade immunotherapy is also a promising treatment to cure cancer. However, their antitumor effects are not sufficient due to some inherent factors. Some studies that combined SDT with immunotherapy or nanoparticles have managed to enhance its efficiency to treat cancers. METHODS In this work, an effective therapeutic strategy that can potentiate the antitumor efficacy of anti-PD-L1 antibody (aPD-L1) is developed by the use of cascade immuno-sonodynamic therapy (immuno-SDT). Titanium dioxide (TiO2), a nanostructured agent for SDT, sonosensitizer Chlorin e6 (Ce6), and immunological adjuvant CpG oligonucleotide (CpG ODN), are used to construct a multifunctional nanosonosensitizer (TiO2-Ce6-CpG). Then, we conducted in vitro and in vivo experiments to explore the antitumor effect of TiO2-Ce6-CpG under ultrasound (US) treatment. RESULTS The characterization tests showed that the nanosonosensitizers are polycrystalline structure with homogeneous sizes, resulting in a good drug loading efficiency. The innovative nanosonosensitizers (TiO2-Ce6-CpG) can not only effectively inhibit tumor growth but also stimulate the immune system to activate the adaptive immune responses, using the TiO2-Ce6 to augment SDT and the immune adjuvant CpG to enhance the immune response. After combined with the aPD-L1, the synergistic effect could not only efficiently inhibit the primary tumor growth but also lead to an inhibition of the non-irradiated pre-existing distant tumors by inducing a strong tumor-specific immune response. CONCLUSION In this study, we present an effective strategy for tumor treatment by combining nanosonosensitizer-augmented SDT and aPD-L1 checkpoint blockade. This work provides a promising strategy and offers a new vision for treating malignant tumors.
Collapse
Affiliation(s)
- Xiaoning Lin
- Department of Neurosurgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Rong Huang
- Department of Child Health, Women and Children’s Hospital, Xiamen University, Xiamen, 361003, People’s Republic of China
| | - Yanlin Huang
- Department of Neurosurgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Kai Wang
- School of Public Health, Xiamen University, Xiamen, 361102, People’s Republic of China
| | - Heng Li
- Department of Neurosurgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Yiheng Bao
- Department of Neurosurgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Chaohui Wu
- Department of Thoracic Surgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Yi Zhang
- Department of Breast Surgery, Xiamen TCM Hospital, Xiamen, 361001, People’s Republic of China
| | - Xinhua Tian
- Department of Neurosurgery, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| | - Xiaomin Wang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital Xiamen University, Xiamen, 361004, People’s Republic of China
| |
Collapse
|
153
|
Jin X, Zeng Q, Zheng J, Xing D, Zhang T. Aptamer-Functionalized Upconverting Nanoformulations for Light-Switching Cancer-Specific Recognition and In Situ Photodynamic-Chemo Sequential Theranostics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9316-9328. [PMID: 33089995 DOI: 10.1021/acsami.0c14730] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Biomarker-activatable theranostic formulations offer the potential for removing specific tumors with a high diagnostic accuracy and a significant pharmacological effect. Herein, we developed a novel activatable theranostic nanoformulation UAS-PD [upconversion nanophosphor (UCNP)-aptamer/ssDNA-pyropheophorbide-a (PPA)-doxyrubicin (DOX)], which can recognize specific cancer cells with sensitivity and trigger the localized photodynamic destruction and enhanced chemotherapy. UAS-PD was constructed by the conjugation of UCNPs and aptamer probes containing the photosensitizer PPA and the chemotherapeutic drug DOX. When cancer cells are present, the UAS-PD specifically binds to PTK7, an overexpressed protein present on the surface of cancer cells, through conformational recombination of the aptamer structure and switches its upconversion luminescence from 655 to 540 nm. This long-lived ratiometric optical signal provides an ultrasensitive detection limit as low as 3.9 nM for PTK7. Changes in the conformation of UAS-PD can also induce PPA to approach UCNPs, which can produce cytotoxic singlet oxygens under near-infrared excitation to destroy the cell membrane and enhance its permeability for the simultaneously released DOX that targets cellular DNA degradation, which results in a highly effective tumor-killing effect by synergistic extra-intracellular sequential damage.
Collapse
Affiliation(s)
- Xudong Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Qin Zeng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Judun Zheng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| |
Collapse
|
154
|
Castro F, Martins C, Silveira MJ, Moura RP, Pereira CL, Sarmento B. Advances on erythrocyte-mimicking nanovehicles to overcome barriers in biological microenvironments. Adv Drug Deliv Rev 2021; 170:312-339. [PMID: 32946921 DOI: 10.1016/j.addr.2020.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
Although nanocarriers offer many advantages as drug delivery systems, their poor stability in circulation, premature drug release and nonspecific uptake in non-target organs have prompted biomimetic approaches using natural cell membranes to camouflage nanovehicles. Among them, erythrocytes, representing the most abundant blood circulating cells, have been extensively investigated for biomimetic coating on artificial nanocarriers due to their upgraded biocompatibility, biodegradability, non-immunogenicity and long-term blood circulation. Due to the cell surface mimetic properties combined with customized core material, erythrocyte-mimicking nanovehicles (EM-NVs) have a wide variety of applications, including drug delivery, imaging, phototherapy, immunomodulation, sensing and detection, that foresee a huge potential for therapeutic and diagnostic applications in several diseases. In this review, we summarize the recent advances in the biomedical applications of EM-NVs in cancer, infection, heart-, autoimmune- and CNS-related disorders and discuss the major challenges and opportunities in this research area.
Collapse
Affiliation(s)
- Flávia Castro
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria José Silveira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Rui Pedro Moura
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
155
|
Yi X, Hu JJ, Dai J, Lou X, Zhao Z, Xia F, Tang BZ. Self-Guiding Polymeric Prodrug Micelles with Two Aggregation-Induced Emission Photosensitizers for Enhanced Chemo-Photodynamic Therapy. ACS NANO 2021; 15:3026-3037. [PMID: 33449627 DOI: 10.1021/acsnano.0c09407] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nowadays, aggregation-induced emission luminogens (AIEgens) with reactive oxygen species (ROS) generating ability have been used as photosensitizers for imaging guided photodynamic therapy (PDT). To achieve enhanced antitumor outcomes, combining AIEgens-based PDT with chemotherapy is an efficient strategy. However, the therapeutic efficiency is hampered by the limited cellular uptake efficiency and the appropriate light irradiation occasion. In this paper, a self-guiding polymeric micelle (TB@PMPT) composed of two AIE photosensitizers and a reduction-sensitive paclitaxel prodrug (PTX-SS-N3) was established for enhanced chemo-photodynamic therapy by a dual-stage light irradiation strategy. When the micelles were accumulated in tumor tissues, the first light irradiation (L1, 6 min) was utilized to facilitate cellular uptake by "photochemical internalization" (PCI). Then, the intracellular glutathione (GSH) would induce the PTX release, micelles disassembly and the aggregation state change of AIEgens. The fluorescence signal change of two AIEgens-based ratiometric fluorescent probe could not only precisely guide the second light irradiation (L2, 18 min) for sufficient ROS production, but also monitor the nonfluorescent drug PTX release in turn. Both in vivo and in vitro studies demonstrated that the dual-stage light irradiation strategy employed for TB@PMPT micelles exhibited a superior therapeutic effect over only 24 min continuous light irradiation.
Collapse
Affiliation(s)
- Xiaoqing Yi
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, China
| | - Jing-Jing Hu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoding Lou
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Zujin Zhao
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
156
|
Xu G, Zhang HX, Li XQ, Yang DC, Liu JY. Red light triggered photodynamic-chemo combination therapy using a prodrug caged by photosensitizer. Eur J Med Chem 2021; 215:113251. [PMID: 33611187 DOI: 10.1016/j.ejmech.2021.113251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/10/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
Development of the drug with high therapeutic efficacy and low toxicity is crucial to cancer ablation. In this study, we have demonstrated a red light-responsive prodrug BDP-TK-CPT by connecting the chemotherapeutic agent camptothecin with a boron dipyrromethene (BDP)-based photosensitizer via a reactive oxygen species (ROS)-labile thioketal chain. Since camptothecin is modified by a BDP-based macrocycle at the active site, the formed prodrug displays an extremely low toxicity in dark. However, upon illumination by red light, it can efficiently generate ROS leading to cell death by photodynamic therapy. Meanwhile, the ROS generated can destroy thioketal group to release free camptothecin which further results in local cell death by chemotherapy. The combined antitumor effects of the prodrug have been verified in HepG2, EC109, and HeLa cancer cells and mice bearing H22 tumors. This study may provide an alternative strategy for stimuli-responsive combination treatment of tumors by conjugation of ROS-activatable prodrugs with photosensitizing agents.
Collapse
Affiliation(s)
- Gan Xu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Hong-Xia Zhang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiao-Qiang Li
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
157
|
Jin J, Wan J, Hu X, Fang T, Ye Z, Wang H. Supramolecular nanoparticles self-assembled from reduction-responsive cabazitaxel prodrugs for effective cancer therapy. Chem Commun (Camb) 2021; 57:2261-2264. [PMID: 33532809 DOI: 10.1039/d0cc06854c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Using hydrophobic cabazitaxel as a target anticancer drug, we show that the conjugation of oligo(ethylene glycol)-oligolactide (OEG-OLA) via a self-immolative linkage induces the self-assembly of the resulting prodrug into injectable nanoparticles. The nanoparticles release chemically unmodified cabazitaxel after endocytosis in cancer cells. With the optimal conjugate, the nanotherapy not only potently induces tumor regression but also has a higher safety margin in animals than the free drug administered in its clinical formulation. Our studies highlight the design rationale that attaching a short amphiphilic oligomer to a toxic drug can convert it to a self-deliverable and safe nanotherapy.
Collapse
Affiliation(s)
- Jiahui Jin
- The First Affiliated Hospital, Zhejiang University School of Medicine, NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, P. R. China.
| | | | | | | | | | | |
Collapse
|
158
|
Bhandari C, Guirguis M, Savan NA, Shrivastava N, Oliveira S, Hasan T, Obaid G. What NIR photodynamic activation offers molecular targeted nanomedicines: Perspectives into the conundrum of tumor specificity and selectivity. NANO TODAY 2021; 36:101052. [PMID: 33552231 PMCID: PMC7864390 DOI: 10.1016/j.nantod.2020.101052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Near infrared (NIR) photodynamic activation is playing increasingly critical roles in cutting-edge anti-cancer nanomedicines, which include spatiotemporal control over induction of therapy, photodynamic priming, and phototriggered immunotherapy. Molecular targeted photonanomedicines (mt-PNMs) are tumor-specific nanoscale drug delivery systems, which capitalize on the unparalleled spatio-temporal precision of NIR photodynamic activation to augment the accuracy of tumor tissue treatment. mt-PNMs are emerging as a paradigm approach for the targeted treatment of solid tumors, yet remain highly complex and multifaceted. While ligand targeted nanomedicines in general suffer from interdependent challenges in biophysics, surface chemistry and nanotechnology, mt-PNMs provide distinct opportunities to synergistically potentiate the effects of ligand targeting. This review provides what we believe to be a much-need demarcation between the processes involved in tumor specificity (biomolecular recognition events) and tumor selectivity (preferential tumor accumulation) of ligand targeted nanomedicines, such as mt-PNMs, and elaborate on what NIR photodynamic activation has to offer. We discuss the interplay between both tumor specificity and tumor selectivity and the degree to which both may play central roles in cutting-edge NIR photoactivable nanotechnologies. A special emphasis is made on NIR photoactivable biomimetic nanotechnologies that capitalize on both specificity and selectivity phenomena to augment the safety and efficacy of photodynamic anti-tumor regimens.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Mina Guirguis
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - N. Anna Savan
- Michigan State University, East Lansing, Michigan, 48824, U.S
| | - Navadeep Shrivastava
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Sabrina Oliveira
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
- Pharmaceutics, Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, U.S
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| |
Collapse
|
159
|
Zhou J, Rao L, Yu G, Cook TR, Chen X, Huang F. Supramolecular cancer nanotheranostics. Chem Soc Rev 2021; 50:2839-2891. [PMID: 33524093 DOI: 10.1039/d0cs00011f] [Citation(s) in RCA: 249] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Among the many challenges in medicine, the treatment and cure of cancer remains an outstanding goal given the complexity and diversity of the disease. Nanotheranostics, the integration of therapy and diagnosis in nanoformulations, is the next generation of personalized medicine to meet the challenges in precise cancer diagnosis, rational management and effective therapy, aiming to significantly increase the survival rate and improve the life quality of cancer patients. Different from most conventional platforms with unsatisfactory theranostic capabilities, supramolecular cancer nanotheranostics have unparalleled advantages in early-stage diagnosis and personal therapy, showing promising potential in clinical translations and applications. In this review, we summarize the progress of supramolecular cancer nanotheranostics and provide guidance for designing new targeted supramolecular theranostic agents. Based on extensive state-of-the-art research, our review will provide the existing and new researchers a foundation from which to advance supramolecular cancer nanotheranostics and promote translationally clinical applications.
Collapse
Affiliation(s)
- Jiong Zhou
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China.
| | | | | | | | | | | |
Collapse
|
160
|
Zhang Y, Ma S, Liu X, Xu Y, Zhao J, Si X, Li H, Huang Z, Wang Z, Tang Z, Song W, Chen X. Supramolecular Assembled Programmable Nanomedicine As In Situ Cancer Vaccine for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007293. [PMID: 33448050 DOI: 10.1002/adma.202007293] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Using nanotechnology for improving the immunotherapy efficiency represents a major research interest in recent years. However, there are paradoxes and obstacles in using a single nanoparticle to fulfill all the requirements in the complicated immune activation processes. Herein, a supramolecular assembled programmable immune activation nanomedicine (PIAN) for sequentially finishing multiple steps after intravenous injection and eliciting robust antitumor immunity in situ is reported. The programmable nanomedicine is constructed by supramolecular assembly via host-guest interactions between poly-[(N-2-hydroxyethyl)-aspartamide]-Pt(IV)/β-cyclodextrin (PPCD), CpG/polyamidoamine-thioketal-adamantane (CpG/PAMAM-TK-Ad), and methoxy poly(ethylene glycol)-thioketal-adamantane (mPEG-TK-Ad). After intravenous injection and accumulation at the tumor site, the high level of reactive oxygen species in the tumor microenvironment promotes PIAN dissociation and the release of PPCD (mediating tumor cell killing and antigen release) and CpG/PAMAM (mediating antigen capturing and transferring to the tumor-draining lymph nodes). This results in antigen-presenting cell activation, antigen presentation, and robust antitumor immune responses. In combination with anti-PD-L1 antibody, the PIAN cures 40% of mice in a colorectal cancer model. This PIAN provides a new framework for designing programmable nanomedicine as in situ cancer vaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
| | - Xinming Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
| | - Yudi Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Hongxiang Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Zhenxin Wang
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Road, Changchun, 130022, China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, 5625 Renmin Road, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| |
Collapse
|
161
|
Luo GF, Chen WH, Zeng X, Zhang XZ. Cell primitive-based biomimetic functional materials for enhanced cancer therapy. Chem Soc Rev 2021; 50:945-985. [PMID: 33226037 DOI: 10.1039/d0cs00152j] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cell primitive-based functional materials that combine the advantages of natural substances and nanotechnology have emerged as attractive therapeutic agents for cancer therapy. Cell primitives are characterized by distinctive biological functions, such as long-term circulation, tumor specific targeting, immune modulation etc. Moreover, synthetic nanomaterials featuring unique physical/chemical properties have been widely used as effective drug delivery vehicles or anticancer agents to treat cancer. The combination of these two kinds of materials will catalyze the generation of innovative biomaterials with multiple functions, high biocompatibility and negligible immunogenicity for precise cancer therapy. In this review, we summarize the most recent advances in the development of cell primitive-based functional materials for cancer therapy. Different cell primitives, including bacteria, phages, cells, cell membranes, and other bioactive substances are introduced with their unique bioactive functions, and strategies in combining with synthetic materials, especially nanoparticulate systems, for the construction of function-enhanced biomaterials are also summarized. Furthermore, foreseeable challenges and future perspectives are also included for the future research direction in this field.
Collapse
Affiliation(s)
- Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | | | | | | |
Collapse
|
162
|
Yuan J, Peng R, Su D, Zhang X, Zhao H, Zhuang X, Chen M, Zhang X, Yuan L. Cell membranes targeted unimolecular prodrug for programmatic photodynamic-chemo therapy. Theranostics 2021; 11:3502-3511. [PMID: 33537100 PMCID: PMC7847693 DOI: 10.7150/thno.55014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/17/2020] [Indexed: 11/29/2022] Open
Abstract
Photodynamic therapy (PDT) has emerged as one of the most up-and-coming non-invasive therapeutic modalities for cancer therapy in rencent years. However, its therapeutic effect was still hampered by the short life span, limited diffusion distance and ineluctable depletion of singlet oxygen (1O2), as well as the hypoxic microenvironment in the tumor tissue. Such problems have limited the application of PDT and appropriate solutions are highly demand. Methods: Herein, a programmatic treatment strategy is proposed for the development of a smart molecular prodrug (D-bpy), which comprise a two-photon photosensitizer and a hypoxia-activated chemotherapeutic prodrug. A rhodamine dye was designed to connect them and track the drug release by the fluorescent signal generated through azo bond cleavage. Results: The prodrug (D-bpy) can stay on the cell membrane and enrich at the tumor site. Upon light irradiation, the therapeutic effect was enhanced by a stepwise treatment: (i) direct generation of 1O2 on the cell membrane induced membrane destruction and promoted the D-bpy uptake; (ii) deep tumor hypoxia caused by two-photon PDT process further triggered the activation of the chemotherapy prodrug. Both in vitro and in vivo experiments, D-bpy have exhabited excellent tumor treatment effect. Conclusion: The innovative programmatic treatment strategy provides new strategy for the design of follow-up anticancer drugs.
Collapse
Affiliation(s)
- Jie Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R China
| | - Rong Peng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R China
| | - Dongdong Su
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Xingxing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R China
| | - Hepeng Zhao
- College of Physics and Microelectronics Science, Hunan University, Changsha 410082, P. R China
| | - Xiujuan Zhuang
- College of Physics and Microelectronics Science, Hunan University, Changsha 410082, P. R China
| | - Mei Chen
- College of Materials Science and Engineering, Hunan University, Changsha 410082, P. R China
| | - Xiaobing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R China
| |
Collapse
|
163
|
Xia J, Pei Q, Zheng M, Xie Z. An activatable fluorescent prodrug of paclitaxel and BODIPY. J Mater Chem B 2021; 9:2308-2313. [DOI: 10.1039/d0tb02510k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A redox-activated paclitaxel prodrug (PTX-S-BDP) was synthesized. PTX-S-BDP NPs were fabricated by the coassembly of PTX-S-BDP with F-127, which can release PTX under redox conditions and exhibit superior cellular imaging and selectivity to cancer cells.
Collapse
Affiliation(s)
- Jinxiu Xia
- School of Chemistry and Life Science
- Advanced Institute of Materials Science
- Changchun University of Technology
- 2055 Yanan Street
- Changchun
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- 5625 Renmin Street
- Changchun
| | - Min Zheng
- School of Chemistry and Life Science
- Advanced Institute of Materials Science
- Changchun University of Technology
- 2055 Yanan Street
- Changchun
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- 5625 Renmin Street
- Changchun
| |
Collapse
|
164
|
Fontana F, Bartolo R, Santos HA. Biohybrid Nanosystems for Cancer Treatment: Merging the Best of Two Worlds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:135-162. [PMID: 33543459 DOI: 10.1007/978-3-030-58174-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
During the last 20+ years, research into the biomedical application of nanotechnology has helped in reshaping cancer treatment. The clinical use of several passively targeted nanosystems resulted in improved quality of care for patients. However, the therapeutic efficacy of these systems is not superior to the original drugs. Moreover, despite extensive investigations into actively targeted nanocarriers, numerous barriers still remain before their successful clinical translation, including sufficient bloodstream circulation time and efficient tumor targeting. The combination of synthetic nanomaterials with biological elements (e.g., cells, cell membranes, and macromolecules) is presently the cutting-edge research in cancer nanotechnology. The features provided by the biological moieties render the particles with prolonged bloodstream circulation time and homotopic targeting to the tumor site. Moreover, cancer cell membranes serve as sources of neoantigens, useful in the formulation of nanovaccines. In this chapter, we will discuss the advantages of biohybrid nanosystems in cancer chemotherapy, immunotherapy, and combined therapy, as well as highlight their preparation methods and clinical translatability.
Collapse
Affiliation(s)
- Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Raquél Bartolo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
165
|
Wang N, Zeng Q, Zhang R, Xing D, Zhang T. Eradication of solid tumors by chemodynamic theranostics with H 2O 2-catalyzed hydroxyl radical burst. Am J Cancer Res 2021; 11:2334-2348. [PMID: 33500728 PMCID: PMC7797687 DOI: 10.7150/thno.49277] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/21/2020] [Indexed: 11/29/2022] Open
Abstract
Activatable theranostics, integrating high diagnostic accuracy and significant therapeutic effect, holds great potential for personalized cancer treatments; however, their chemodynamic modality is rarely exploited. Herein, we report a new in situ activatable chemodynamic theranostics PAsc/Fe@Cy7QB to specifically recognize and eradicate cancer cells with H2O2-catalyzed hydroxyl radical (•OH) burst cascade. Methods: The nanomicelles PAsc/Fe@Cy7QB were constructed by self-assembly of acid-responsive copolymers incorporating ascorbates and acid-sensitive Schiff base-Fe2+ complexes as well as H2O2-responsive adjuvant Cy7QB. Results: Upon systematic delivery of PAsc/Fe@Cy7QB into cancer cells, the acidic microenvironment triggered disassembly of the nanomicelles. The released Fe2+ catalyzed the oxidation of ascorbate monoanion (AscH-) to efficiently produce H2O2. The released H2O2, together with the endogenous H2O2, could be converted into highly active •OH via the Fenton reaction, resulting in enhanced Fe-mediated T1 magnetic resonance imaging (MRI). The synchronously released Cy7QB was activated by H2O2 to produce a glutathione (GSH)-scavenger quinone methide to boost the •OH yield and recover the Cy7 dye for fluorescence and photoacoustic imaging. Conclusion: The biodegradable PAsc/Fe@Cy7QB designed for tumor-selective multimodal imaging and high therapeutic effect provides an exemplary paradigm for precise chemodynamic theranostic.
Collapse
|
166
|
Tiwari R, Shinde PS, Sreedharan S, Dey AK, Vallis KA, Mhaske SB, Pramanik SK, Das A. Photoactivatable prodrug for simultaneous release of mertansine and CO along with a BODIPY derivative as a luminescent marker in mitochondria: a proof of concept for NIR image-guided cancer therapy. Chem Sci 2020; 12:2667-2673. [PMID: 34164035 PMCID: PMC8179275 DOI: 10.1039/d0sc06270g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Controlled and efficient activation is the crucial aspect of designing an effective prodrug. Herein we demonstrate a proof of concept for a light activatable prodrug with desired organelle specificity. Mertansine, a benzoansamacrolide, is an efficient microtubule-targeting compound that binds at or near the vinblastine-binding site in the mitochondrial region to induce mitotic arrest and cell death through apoptosis. Despite its efficacy even in the nanomolar level, this has failed in stage 2 of human clinical trials owing to the lack of drug specificity and the deleterious systemic toxicity. To get around this problem, a recent trend is to develop an antibody-conjugatable maytansinoid with improved tumor/organelle-specificity and lesser systematic toxicity. Endogenous CO is recognized as a regulator of cellular function and for its obligatory role in cell apoptosis. CO blocks the proliferation of cancer cells and effector T cells, and the primary target is reported to be the mitochondria. We report herein a new mitochondria-specific prodrug conjugate (Pro-DC) that undergoes a photocleavage reaction on irradiation with a 400 nm source (1.0 mW cm−2) to induce a simultaneous release of the therapeutic components mertansine and CO along with a BODIPY derivative (BODIPY(PPH3)2) as a luminescent marker in the mitochondrial matrix. The efficacy of the process is demonstrated using MCF-7 cells and could effectively be visualized by probing the intracellular luminescence of BODIPY(PPH3)2. This provides a proof-of-concept for designing a prodrug for image-guided combination therapy for mainstream treatment of cancer. Simultaneous release of two therapeutic reagents, mertansine and CO through photo-induced cleavage of a mitochondria-specific prodrug with improved drug efficacy.![]()
Collapse
Affiliation(s)
- Rajeshwari Tiwari
- Central Salt and Marine Chemicals Research Institute Bhavnagar Gujarat India .,Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | | | - Sreejesh Sreedharan
- Oxford Institute for Radiation Oncology, University of Oxford Oxford OX3 7DQ UK
| | - Anik Kumar Dey
- Central Salt and Marine Chemicals Research Institute Bhavnagar Gujarat India .,Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Katherine A Vallis
- Oxford Institute for Radiation Oncology, University of Oxford Oxford OX3 7DQ UK
| | - Santosh B Mhaske
- CSIR-National Chemical Laboratory Pune 411008 India .,Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sumit Kumar Pramanik
- Central Salt and Marine Chemicals Research Institute Bhavnagar Gujarat India .,Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Amitava Das
- Indian Institute of Science Education and Research Kolkata Mohanpur 741246 West Bengal India
| |
Collapse
|
167
|
Weinstain R, Slanina T, Kand D, Klán P. Visible-to-NIR-Light Activated Release: From Small Molecules to Nanomaterials. Chem Rev 2020; 120:13135-13272. [PMID: 33125209 PMCID: PMC7833475 DOI: 10.1021/acs.chemrev.0c00663] [Citation(s) in RCA: 339] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Photoactivatable (alternatively, photoremovable, photoreleasable, or photocleavable) protecting groups (PPGs), also known as caged or photocaged compounds, are used to enable non-invasive spatiotemporal photochemical control over the release of species of interest. Recent years have seen the development of PPGs activatable by biologically and chemically benign visible and near-infrared (NIR) light. These long-wavelength-absorbing moieties expand the applicability of this powerful method and its accessibility to non-specialist users. This review comprehensively covers organic and transition metal-containing photoactivatable compounds (complexes) that absorb in the visible- and NIR-range to release various leaving groups and gasotransmitters (carbon monoxide, nitric oxide, and hydrogen sulfide). The text also covers visible- and NIR-light-induced photosensitized release using molecular sensitizers, quantum dots, and upconversion and second-harmonic nanoparticles, as well as release via photodynamic (photooxygenation by singlet oxygen) and photothermal effects. Release from photoactivatable polymers, micelles, vesicles, and photoswitches, along with the related emerging field of photopharmacology, is discussed at the end of the review.
Collapse
Affiliation(s)
- Roy Weinstain
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Tomáš Slanina
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czech Republic
| | - Dnyaneshwar Kand
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Petr Klán
- Department
of Chemistry and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
168
|
Yang Y, Zeng W, Huang P, Zeng X, Mei L. Smart materials for drug delivery and cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200042] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yao Yang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Weiwei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Ping Huang
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Xiaowei Zeng
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
| | - Lin Mei
- Institute of Pharmaceutics School of Pharmaceutical Sciences (Shenzhen) Sun Yat‐sen University Shenzhen China
- Tianjin Key Laboratory of Biomedical Materials Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy Institute of Biomedical Engineering Chinese Academy of Medical Sciences & Peking Union Medical College Tianjin China
| |
Collapse
|
169
|
Advanced engineered nanoparticulate platforms to address key biological barriers for delivering chemotherapeutic agents to target sites. Adv Drug Deliv Rev 2020; 167:170-188. [PMID: 32622022 DOI: 10.1016/j.addr.2020.06.030] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
The widespread development of nanocarriers to deliver chemotherapeutics to specific tumor sites has been motivated by the lack of selective targeting during chemotherapy inducing serious side effects and low therapeutic efficacy. The utmost challenge in targeted cancer therapies is the ineffective drug delivery system, in which the drug-loaded nanocarriers are hindered by multiple complex biological barriers that compromise the therapeutic efficacy. Despite considerable progress engineering novel nanoplatforms for the delivery of chemotherapeutics, there has been limited success in a clinical setting. In this review, we identify and analyze design strategies for improved therapeutic efficacy and unique properties of nanoplatforms, including liposomes, polymeric micelles, nanogels, and dendrimers. We provide a comprehensive and integral description of key biological barriers that nanoplatforms are exposed to during their in vivo journey and discuss associated strategies to overcome these barriers based on the latest research and information available in the field. We expect this review to provide constructive information for the rational design of more effective nanoplatforms to advance precision therapies and accelerate their clinical translation.
Collapse
|
170
|
Tao Y, Chan HF, Shi B, Li M, Leong KW. Light: A Magical Tool for Controlled Drug Delivery. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2005029. [PMID: 34483808 PMCID: PMC8415493 DOI: 10.1002/adfm.202005029] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Indexed: 05/04/2023]
Abstract
Light is a particularly appealing tool for on-demand drug delivery due to its noninvasive nature, ease of application and exquisite temporal and spatial control. Great progress has been achieved in the development of novel light-driven drug delivery strategies with both breadth and depth. Light-controlled drug delivery platforms can be generally categorized into three groups: photochemical, photothermal, and photoisomerization-mediated therapies. Various advanced materials, such as metal nanoparticles, metal sulfides and oxides, metal-organic frameworks, carbon nanomaterials, upconversion nanoparticles, semiconductor nanoparticles, stimuli-responsive micelles, polymer- and liposome-based nanoparticles have been applied for light-stimulated drug delivery. In view of the increasing interest in on-demand targeted drug delivery, we review the development of light-responsive systems with a focus on recent advances, key limitations, and future directions.
Collapse
Affiliation(s)
- Yu Tao
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingyang Shi
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Kam W Leong
- Department of Biomedical Engineering, Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
171
|
Yang B, Gao J, Pei Q, Xu H, Yu H. Engineering Prodrug Nanomedicine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002365. [PMID: 33304763 PMCID: PMC7709995 DOI: 10.1002/advs.202002365] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has shifted the clinical paradigm of cancer management. However, despite promising initial progress, immunotherapeutic approaches to cancer still suffer from relatively low response rates and the possibility of severe side effects, likely due to the low inherent immunogenicity of tumor cells, the immunosuppressive tumor microenvironment, and significant inter- and intratumoral heterogeneity. Recently, nanoformulations of prodrugs have been explored as a means to enhance cancer immunotherapy by simultaneously eliciting antitumor immune responses and reversing local immunosuppression. Prodrug nanomedicines, which integrate engineering advances in chemistry, oncoimmunology, and material science, are rationally designed through chemically modifying small molecule drugs, peptides, or antibodies to yield increased bioavailability and spatiotemporal control of drug release and activation at the target sites. Such strategies can help reduce adverse effects and enable codelivery of multiple immune modulators to yield synergistic cancer immunotherapy. In this review article, recent advances and translational challenges facing prodrug nanomedicines for cancer immunotherapy are overviewed. Last, key considerations are outlined for future efforts to advance prodrug nanomedicines aimed to improve antitumor immune responses and combat immune tolerogenic microenvironments.
Collapse
Affiliation(s)
- Bin Yang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Qing Pei
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
172
|
Lin C, Tong F, Liu R, Xie R, Lei T, Chen Y, Yang Z, Gao H, Yu X. GSH-responsive SN38 dimer-loaded shape-transformable nanoparticles with iRGD for enhancing chemo-photodynamic therapy. Acta Pharm Sin B 2020; 10:2348-2361. [PMID: 33354506 PMCID: PMC7745177 DOI: 10.1016/j.apsb.2020.10.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/12/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Accurate tumor targeting, deep penetration and superb retention are still the main pursuit of developing excellent nanomedicine. To achieve these requirements, a stepwise stimuli-responsive strategy was developed through co-administration tumor penetration peptide iRGD with shape-transformable and GSH-responsive SN38-dimer (d-SN38)-loaded nanoparticles (d-SN38@NPs/iRGD). Upon intravenous injection, d-SN38@NPs with high drug loading efficiency (33.92 ± 1.33%) could effectively accumulate and penetrate into the deep region of tumor sites with the assistance of iRGD. The gathered nanoparticles simultaneously transformed into nanofibers upon 650 nm laser irradiation at tumor sites so as to promote their retention in the tumor and burst release of reactive oxygen species for photodynamic therapy. The loaded d-SN38 with disulfide bond responded to the high level of GSH in tumor cytoplasm, which consequently resulted in SN38 release and excellent chemo-photodynamic effect on tumor. In vitro, co-administering iRGD with d-SN38@NPs+laser showed higher cellular uptake, apoptosis ratio and multicellular spheroid penetration. In vivo, d-SN38@NPs/iRGD+laser displayed advanced penetration and accumulation in tumor, leading to 60.89% of tumor suppression in 4T1 tumor-bearing mouse model with a favorable toxicity profile. Our new strategy combining iRGD with structural transformable nanoparticles greatly improves tumor targeting, penetrating and retention, and empowers anticancer efficacy.
Collapse
Affiliation(s)
- Congcong Lin
- Department of Radiology, Zhuhai People's Hospital, Jinan University, Zhuhai 519000, China
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Fan Tong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Rui Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Rou Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Ting Lei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Yuxiu Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Zhihang Yang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Sichuan 610041, China
- Corresponding authors. Tel./fax: +86 28 85502532.
| | - Xiangrong Yu
- Department of Radiology, Zhuhai People's Hospital, Jinan University, Zhuhai 519000, China
- Corresponding authors. Tel./fax: +86 28 85502532.
| |
Collapse
|
173
|
Huang L, Zhao S, Fang F, Xu T, Lan M, Zhang J. Advances and perspectives in carrier-free nanodrugs for cancer chemo-monotherapy and combination therapy. Biomaterials 2020; 268:120557. [PMID: 33260095 DOI: 10.1016/j.biomaterials.2020.120557] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/09/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Nanocarrier-based drug delivery systems hold impressive promise for biomedical application because of their excellent water dispersibility, prolonged blood circulation time, increased drug accumulation in tumors, and potential in combination therapeutics. However, most nanocarriers suffer from low drug-loading efficiency, poor therapeutic effectiveness, potential systematic toxicity, and unstable metabolism. As an alternative, carrier-free nanodrugs, completely formulated with one or more drugs, have attracted increasing attention in cancer therapy due to their advantage of improved pharmacodynamics/pharmacokinetics, reduced toxicity, and high drug-loading. In recent years, carrier-free nanodrugs have contributed to progress in a variety of therapeutic modalities. In this review, different common strategies for carrier-free nanodrugs preparation are first summarized, mainly including nanoprecipitation, template-assisted nanoprecipitation, thin-film hydration, spray-drying technique, supercritical fluid (SCF) technique, and wet media milling. Then we describe the recently reported carrier-free nanodrugs for cancer chemo-monotherapy or combination therapy. The advantages of anti-cancer drugs combined with other chemotherapeutic, photosensitizers, photothermal, immunotherapeutic or gene drugs have been demonstrated. Finally, a future perspective is introduced to highlight the existing challenges and possible solutions toward clinical application of currently developed carrier-free nanodrugs, which may be instructive to the design of effective carrier-free regimens in the future.
Collapse
Affiliation(s)
- Li Huang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Shaojing Zhao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Ting Xu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Minhuan Lan
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China.
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, PR China.
| |
Collapse
|
174
|
Zhao C, Chen J, Zhong R, Chen DS, Shi J, Song J. Materialien mit Selektivität für oxidative Molekülspezies für die Diagnostik und Therapie. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201915833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Caiyan Zhao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jingxiao Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Pharmaceutical Sciences Jiangnan University Wuxi 214122 PR China
| | - Ruibo Zhong
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Dean Shuailin Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jinjun Shi
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
| |
Collapse
|
175
|
Zhao C, Chen J, Zhong R, Chen DS, Shi J, Song J. Oxidative‐Species‐Selective Materials for Diagnostic and Therapeutic Applications. Angew Chem Int Ed Engl 2020; 60:9804-9827. [DOI: 10.1002/anie.201915833] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/15/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Caiyan Zhao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jingxiao Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Pharmaceutical Sciences Jiangnan University Wuxi 214122 PR China
| | - Ruibo Zhong
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Dean Shuailin Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jinjun Shi
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
| |
Collapse
|
176
|
Zuo S, Sun B, Yang Y, Zhou S, Zhang Y, Guo M, Sun M, Luo C, He Z, Sun J. Probing the Superiority of Diselenium Bond on Docetaxel Dimeric Prodrug Nanoassemblies: Small Roles Taking Big Responsibilities. SMALL 2020; 16:e2005039. [PMID: 33078579 DOI: 10.1002/smll.202005039] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Indexed: 02/05/2023]
Abstract
The current state of chemotherapy is far from satisfaction, restricted by the inefficient drug delivery and the off-target toxicity. Prodrug nanoassemblies are emerging as efficient platforms for chemotherapy. Herein, three docetaxel dimeric prodrugs are designed using diselenide bond, disulfide bond, or dicarbide bond as linkages. Interestingly, diselenide bond-bridged dimeric prodrug can self-assemble into stable nanoparticles with impressive high drug loading (≈70%, w/w). Compared with disulfide bond and dicarbide bond, diselenide bond greatly facilitates the self-assembly of dimeric prodrug, and then improves the colloidal stability, blood circulation time, and antitumor efficacy of prodrug nanoassemblies. Furthermore, the redox-sensitive diselenide bond can specifically respond to the overexpressed reactive oxygen species and glutathione in tumor cells, leading to tumor-specific drug release. Therefore, diselenide bond bridged prodrug nanoassemblies exhibit discriminating cytotoxicity between tumor cells and normal cells, significantly alleviating the systemic toxicity of docetaxel. The present work gains in-depth insight into the impact of diselenide bond on the dimeric prodrug nanoassemblies, and provides promising strategies for the rational design of the high efficiency-low toxicity chemotherapeutical nanomedicines.
Collapse
Affiliation(s)
- Shiyi Zuo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Yinxian Yang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Shuang Zhou
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Yu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Mengran Guo
- Department of Clinical Pharmacy, West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Mengchi Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| |
Collapse
|
177
|
Zhou S, Hu X, Xia R, Liu S, Pei Q, Chen G, Xie Z, Jing X. A Paclitaxel Prodrug Activatable by Irradiation in a Hypoxic Microenvironment. Angew Chem Int Ed Engl 2020; 59:23198-23205. [PMID: 32852145 DOI: 10.1002/anie.202008732] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/13/2020] [Indexed: 12/15/2022]
Abstract
The innate hypoxic microenvironment of most solid tumors has a major influence on tumor growth, invasiveness, and distant metastasis. Here, a hypoxia-activated self-immolative prodrug of paclitaxel (PTX2 -Azo) was synthesized and encapsulated by a peptide copolymer decorated with the photosensitizer chlorin e6 (Ce6) to prepare light-boosted PTX nanoparticle (Ce6/PTX2 -Azo NP). In this nanoparticle, PTX2 -Azo prevents premature drug leakage and realizes specific release in hypoxic tumor microenvironment and the photosensitizer Ce6 not only efficiently generates singlet oxygen under light irradiation but also acts as a positive amplifier to promote the release of PTX. The combination of photodynamic therapy (PDT) and chemotherapy results in excellent antitumor efficacy, demonstrating the great potential for synergistic cancer therapy.
Collapse
Affiliation(s)
- Shiyu Zhou
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Rui Xia
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Shi Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Guang Chen
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| |
Collapse
|
178
|
Zhou S, Hu X, Xia R, Liu S, Pei Q, Chen G, Xie Z, Jing X. A Paclitaxel Prodrug Activatable by Irradiation in a Hypoxic Microenvironment. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shiyu Zhou
- Department of Thyroid Surgery The First Hospital of Jilin University Changchun Jilin 130021 China
| | - Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Rui Xia
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Shi Liu
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Guang Chen
- Department of Thyroid Surgery The First Hospital of Jilin University Changchun Jilin 130021 China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| |
Collapse
|
179
|
Karaosmanoglu S, Zhou M, Shi B, Zhang X, Williams GR, Chen X. Carrier-free nanodrugs for safe and effective cancer treatment. J Control Release 2020; 329:805-832. [PMID: 33045313 DOI: 10.1016/j.jconrel.2020.10.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
Clinical applications of many anti-cancer drugs are restricted due to their hydrophobic nature, requiring use of harmful organic solvents for administration, and poor selectivity and pharmacokinetics resulting in off-target toxicity and inefficient therapies. A wide variety of carrier-based nanoparticles have been developed to tackle these issues, but such strategies often fail to encapsulate drug efficiently and require significant amounts of inorganic and/or organic nanocarriers which may cause toxicity problems in the long term. Preparation of nano-formulations for the delivery of water insoluble drugs without using carriers is thus desired, requiring elegantly designed strategies for products with high quality, stability and performance. These strategies include simple self-assembly or involving chemical modifications via coupling drugs together or conjugating them with various functional molecules such as lipids, carbohydrates and photosensitizers. During nanodrugs synthesis, insertion of redox-responsive linkers and tumor targeting ligands endows them with additional characteristics like on-target delivery, and conjugation with immunotherapeutic reagents enhances immune response alongside therapeutic efficacy. This review aims to summarize the methods of making carrier-free nanodrugs from hydrophobic drug molecules, evaluating their performance, and discussing the advantages, challenges, and future development of these strategies.
Collapse
Affiliation(s)
- Sena Karaosmanoglu
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK
| | - Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Bingyang Shi
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xiujuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, PR China.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK.
| |
Collapse
|
180
|
Spanjers JM, Städler B. Cell Membrane Coated Particles. ACTA ACUST UNITED AC 2020; 4:e2000174. [DOI: 10.1002/adbi.202000174] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/14/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Järvi M. Spanjers
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 Aarhus C 8000 Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University Gustav Wieds Vej 14 Aarhus C 8000 Denmark
| |
Collapse
|
181
|
Liu D, Shu G, Jin F, Qi J, Xu X, Du Y, Yu H, Wang J, Sun M, You Y, Zhu M, Chen M, Zhu L, Shen Q, Ying X, Lou X, Jiang S, Du Y. ROS-responsive chitosan-SS31 prodrug for AKI therapy via rapid distribution in the kidney and long-term retention in the renal tubule. SCIENCE ADVANCES 2020; 6:6/41/eabb7422. [PMID: 33036968 PMCID: PMC7546709 DOI: 10.1126/sciadv.abb7422] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 08/21/2020] [Indexed: 05/06/2023]
Abstract
The development of drugs with rapid distribution in the kidney and long-term retention in the renal tubule is a breakthrough for enhanced treatment of acute kidney injury (AKI). Here, l-serine-modified chitosan (SC) was synthesized as a potential AKI kidney-targeting agent due to the native cationic property of chitosan and specific interaction between kidney injury molecule-1 (Kim-1) and serine. Results indicated that SC was rapidly accumulated and long-term retained in ischemia-reperfusion-induced AKI kidneys, especially in renal tubules, which was possibly due to the specific interactions between SC and Kim-1. SC-TK-SS31 was then prepared by conjugating SS31, a mitochondria-targeted antioxidant, to SC via reactive oxygen species (ROS)-sensitive thioketal linker. Because of the effective renal distribution combined with ROS-responsive drug release behavior, the administration of SC-TK-SS31 led to an enhanced therapeutic effect of SS31 by protecting mitochondria from damage and reducing the oxidative stress, inflammation, and cell apoptosis.
Collapse
Affiliation(s)
- Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Gaofeng Shu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Feiyang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Jing Qi
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiaoling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yan Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Hui Yu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Jun Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Mingchen Sun
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yuchan You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Minxia Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Meixuan Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Luwen Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Qiying Shen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiaoying Ying
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xuefang Lou
- School of Medicine, Zhejiang University City College, Hangzhou 310015, China.
| | - Saiping Jiang
- Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China.
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China.
| |
Collapse
|
182
|
Peng H, Xu Z, Wang Y, Feng N, Yang W, Tang J. Biomimetic Mesoporous Silica Nanoparticles for Enhanced Blood Circulation and Cancer Therapy. ACS APPLIED BIO MATERIALS 2020; 3:7849-7857. [DOI: 10.1021/acsabm.0c01014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Haibao Peng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhuoyuan Xu
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, United States
| | - Yongcheng Wang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jing Tang
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
183
|
Li Y, Tan X, Liu X, Liu L, Fang Y, Rao R, Ren Y, Yang X, Liu W. Enhanced anticancer effect of doxorubicin by TPGS-coated liposomes with Bcl-2 siRNA-corona for dual suppression of drug resistance. Asian J Pharm Sci 2020; 15:646-660. [PMID: 33193866 PMCID: PMC7610212 DOI: 10.1016/j.ajps.2019.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/30/2019] [Accepted: 10/09/2019] [Indexed: 12/21/2022] Open
Abstract
Multiple drug resistance (MDR) is a tough problem in developing hepatocellular carcinoma (HCC) therapy. Here, we developed TPGS-coated cationic liposomes with Bcl-2 siRNA corona to load doxorubicin (Dox) i.e., Bcl-2 siRNA/Dox-TPGS-LPs, to enhance anticancer effect of Dox in HCC-MDR. TPGS i.e., d-α-tocopheryl polyethylene glycol 1000 succinate, inhibited P-glycoprotein (P-gp) efflux pump and Bcl-2 siRNA suppressed anti-apoptotic Bcl-2 protein. The Bcl-2 siRNA loaded in the liposomal corona was observed under transmission electron microscopy. The stability and hemolysis evaluation demonstrated Bcl-2 siRNA/Dox-TPGS-LPs had good biocompatibility and siRNA-corona could protect the liposomal core to avoid the attachment of fetal bovine serum. In drug-resistant cells, TPGS effectively prolonged intracellular Dox retention time and siRNA-corona did improve the internalization of Dox from liposomes. In vitro and in vivo anticancer effect of this dual-functional nanostructure was examined in HCC-MDR Bel7402/5-FU tumor model. MTT assay confirmed the IC50 value of Dox was 20-50 fold higher in Bel7402/5-FU MDR cells than that in sensitive Bel7402 cells. Bcl-2 siRNA corona successfully entered the cytosol of Bel7402/5-FU MDR cells to downregulate Bcl-2 protein levels in vitro and in vivo. Bcl-2 siRNA/Dox-TPGS-LPs showed superior to TPGS- (or siRNA-) linked Dox liposomes in cell apoptosis and cytotoxicity assay in Bel7402/5-FU MDR cells, and 7-fold greater effect than free Dox in tumor growth inhibition of Bel7402/5-FU xenograft nude mice. In conclusion, TPGS-coated cationic liposomes with Bcl-2 siRNA corona had the capacity to inhibit MDR dual-pathways and subsequently improved the anti-tumor activity of the chemotherapeutic agent co-delivered to a level that cannot be achieved by inhibiting a MDR single way.
Collapse
Affiliation(s)
- Yinghuan Li
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing 100069, China
| | - Xi Tan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xuhan Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lingyan Liu
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing 100069, China
| | - Yan Fang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Rong Rao
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuanyuan Ren
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wei Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
184
|
Huang L, Chen X, Bian Q, Zhang F, Wu H, Wang H, Gao J. Photosensitizer-stabilized self-assembling nanoparticles potentiate chemo/photodynamic efficacy of patient-derived melanoma. J Control Release 2020; 328:325-338. [PMID: 32889052 DOI: 10.1016/j.jconrel.2020.08.062] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/13/2022]
Abstract
Development of injectable nanoparticles for delivery of active anticancer compounds often requires complicated schemes that involve tedious synthetic protocols and nanoformulations. In particular, clinical translation of synergistic nanoparticles that can facilitate multimodal therapies remains a considerable challenge. Herein, we describe a self-assembling, small-molecule nanosystem with unique properties, including near-infrared (NIR) light-responsive drug activation, size transformability, combinatorial synergy, and substantially reduced toxicity. Ligation of anticancer cabazitaxel (CTX) drugs via a reactive oxygen species-activatable thioketal linkage generates a dimeric TKdC prodrug, and subsequent coassembly with a photosensitizer, chlorin e6 (Ce6), forms colloidal-stable nanoassemblies (termed psTKdC NAs). Upon NIR laser irradiation, psTKdC NAs are transformed into smaller size particles and facilitate production of pharmacologically active CTX. Importantly, reactive oxygen species yielded by coassembled Ce6 can synergize with chemotherapy to achieve potent combinatorial effects. In a preclinical orthotopic model of an aggressive, human melanoma patient-derived xenograft (PDX), we show that administration of psTKdC NAs followed by laser irradiation produced durable tumor regression, with the tumors being completely eradicated in three of six PDXs. Furthermore, low systemic toxicity of this smart, photo-activatable nanotherapy was observed in animals. The new self-deliverable combinatorial system addresses essential requirements for high efficacy, safety, and translational capacity and deserves further investigation.
Collapse
Affiliation(s)
- Lingling Huang
- Institute of Pharmaceutics, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou 310058, PR China; The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Zhejiang, Hangzhou 310058, PR China
| | - Xiaona Chen
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Zhejiang, Hangzhou 310058, PR China
| | - Qiong Bian
- Institute of Pharmaceutics, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou 310058, PR China
| | - Fu Zhang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Zhejiang, Hangzhou 310058, PR China
| | - Honghui Wu
- Institute of Pharmaceutics, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou 310058, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Zhejiang, Hangzhou 310058, PR China.
| | - Jianqing Gao
- Institute of Pharmaceutics, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, Hangzhou 310058, PR China.
| |
Collapse
|
185
|
Liu C, Liu Q, Chen L, Li M, Yin J, Zhu X, Chen D. A pH-Sensitive Self-Assembled and Carrier-Free Nanoparticle Based on Charge Reversal for Enhanced Synergetic Chemo-Phototherapy. Adv Healthc Mater 2020; 9:e2000899. [PMID: 33448702 DOI: 10.1002/adhm.202000899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Indexed: 12/21/2022]
Abstract
To overcome biological barriers for nanoparticles (NPs) efficaciously accumulated at tumor sites, as well as enhancing the performance of drug delivery systems, a carrier-free nanoparticle based on charge reversal is designed for improved synergetic chemo-phototherapy for cancer treatment. In this system, doxorubicin (Dox) and zinc phthalocyanine (ZnPc) are self-assembled through noncovalent interactions (π-π stacking, hydrophobic forces) to avoid the possible toxicity of excipient, complex chemical conjugations and batch-to-batch variation. A trace amount of poly(2-(di-methylamino) ethylmethacrylate)- poly[(R)-3-hydroxybutyrate]- poly(2-(dimethylamino) ethylmethacrylate (PDMAEMA-PHB-PDMAEMA) is modified on the surface of Dox-ZnPc to construct the novel nanoparticles, namely DZP, with long-term stability, and with a dual-drug load content of up to ≈90%. The drug delivery system (DDS) can effectively decrease its toxicity among physical circulation and increase the accumulation at the tumor site. Moreover, the developed DZP nanoparticles show excellent photo-chemotherapy, photoacoustic (PA) and fluorescence (FL) imaging characteristics for multimodal imaging-guided synergetic therapy.
Collapse
Affiliation(s)
- Chen Liu
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Qiuhong Liu
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Luping Chen
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Mao Li
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Jieli Yin
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Xuan Zhu
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| | - Dengyue Chen
- School of Pharmaceutical Sciences Xiamen University Xiamen Fujian 361102 China
| |
Collapse
|
186
|
Yang B, Wei L, Wang Y, Li N, Ji B, Wang K, Zhang X, Zhang S, Zhou S, Yao X, Song H, Wu Y, Zhang H, Kan Q, Jin T, Sun J. Oxidation-strengthened disulfide-bridged prodrug nanoplatforms with cascade facilitated drug release for synergetic photochemotherapy. Asian J Pharm Sci 2020; 15:637-645. [PMID: 33193865 PMCID: PMC7610204 DOI: 10.1016/j.ajps.2019.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/05/2019] [Accepted: 09/13/2019] [Indexed: 01/17/2023] Open
Abstract
One of the major barriers in utilizing prodrug nanocarriers for cancer therapy is the slow release of parent drug in tumors. Tumor cells generally display the higher oxidative level than normal cells, and also displayed the heterogeneity in terms of redox homeostasis level. We previously found that the disulfide bond-linkage demonstrates surprising oxidation-sensitivity to form the hydrophilic sulfoxide and sulphone groups. Herein, we develop oxidation-strengthened prodrug nanosystem loaded with pyropheophorbide a (PPa) to achieve light-activatable cascade drug release and enhance therapeutic efficacy. The disulfide bond-driven prodrug nanosystems not only respond to the redox-heterogeneity in tumor, but also respond to the exogenous oxidant (singlet oxygen) elicited by photosensitizers. Once the prodrug nanoparticles (NPs) are activated under irradiation, they would undergo an oxidative self-strengthened process, resulting in a facilitated drug cascade release. The IC50 value of the PPa@PTX-S-S NPs without irradiation was 2-fold higher than those of NPs plus irradiation. In vivo, the PPa@PTX prodrug NPs display prolonged systemic circulation and increased accumulation in tumor site. The PPa@PTX-S-S NPs showed much higher efficiency than free PTX or the PPa@PTX-C-C NPs to suppress the growth of 4T1 tumors. Therefore, this novel oxidation-strengthened disulfide-bridged prodrug-nanosystem has a great potential in the enhanced efficacy of cancer synergetic photochemotherapy.
Collapse
Affiliation(s)
- Bin Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Lin Wei
- Ministry of Education, Heilongjiang University, Harbin 150500, China
- Sino-Russian Joint Graduate School, Heilongjiang University, Harbin 150080, China
| | - Yuequan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Na Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bin Ji
- Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Kaiyuan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuanbo Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shenwu Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohui Yao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hang Song
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yusheng Wu
- Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiming Kan
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Jin
- Ministry of Education, Heilongjiang University, Harbin 150500, China
- School of Life Sciences, Heilongjiang University, Harbin 150080, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
187
|
Harnessing cells to deliver nanoparticle drugs to treat cancer. Biotechnol Adv 2020; 42:107339. [DOI: 10.1016/j.biotechadv.2019.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/27/2022]
|
188
|
Tong X, Zhang L, Li L, Li Y, Yang Z, Zhu D, Xie Z. Water-soluble cyclometalated Ir(III) complexes as carrier-free and pure nanoparticle photosensitizers for photodynamic therapy and cell imaging. Dalton Trans 2020; 49:11493-11497. [PMID: 32789382 DOI: 10.1039/d0dt02584d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Herein, we provide a new and facile strategy to successfully overcome the inherent aggregation-caused quenching effect and hydrophobicity that exist in traditional PSs by the introduction of sodium salts. The obtained water-soluble Ir(iii) complexes as carrier-free and pure nanoparticle photosensitizers exhibit excellent performance in photodynamic therapy and cell imaging.
Collapse
Affiliation(s)
- Xiaofan Tong
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province 130024, P. R. China.
| | - Liping Zhang
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province 130024, P. R. China.
| | - Lijuan Li
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province 130024, P. R. China.
| | - Yite Li
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, P. R. China.
| | - Zhiyu Yang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, P. R. China.
| | - Dongxia Zhu
- Key Laboratory of Nanobiosensing and Nanobioanalysis at Universities of Jilin Province, Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province 130024, P. R. China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, P. R. China.
| |
Collapse
|
189
|
Chen R, Chen WC, Yan L, Tian S, Liu B, Chen X, Lee CS, Zhang W. Harnessing combinational phototherapy via post-synthetic PpIX conjugation on nanoscale metal-organic frameworks. J Mater Chem B 2020; 7:4763-4770. [PMID: 31389960 DOI: 10.1039/c9tb01154d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanomaterial-mediated phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is an effective anticancer intervention that relies on light activation of photoactive nanomaterials localized in tumors. Recently, combinational PDT/PTT offered a practical pathway to relieve resistance of monotherapy, surmount undesirable side effects and provide a synergistic effect to enhance phototherapeutic efficiency. Herein, we report a facile strategy to integrate protoporphyrin IX (PpIX) into nanoscale metal-organic frameworks (NMOFs) and control their photoactive properties for combinational cancer PDT and PTT. With optimized PpIX conjugation, the as-fabricated nanoparticles (nPCU NPs) exhibit (1) improved dispersibility and particle stability, (2) simultaneous generation of reactive oxygen species and heat effectively through activation by a single-wavelength laser of 635 nm, and (3) maintenance of porosity for further application as drug delivery vehicles. Moreover, in vitro investigation of nPCU NPs demonstrates effective cellular uptake, successful endosomal escape and enhanced phototherapeutic efficacy under both normoxic and hypoxic conditions. Therefore, this study developed a novel type of phototherapeutic nanoplatform with optimal properties for applicable cancer phototherapy.
Collapse
Affiliation(s)
- Rui Chen
- Center of Super-Diamond and Advanced Films (COSDAF), Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
190
|
Wang X, Xuan Z, Zhu X, Sun H, Li J, Xie Z. Near-infrared photoresponsive drug delivery nanosystems for cancer photo-chemotherapy. J Nanobiotechnology 2020; 18:108. [PMID: 32746846 PMCID: PMC7397640 DOI: 10.1186/s12951-020-00668-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022] Open
Abstract
Drug delivery systems (DDSs) based on nanomaterials have shown a promise for cancer chemotherapy; however, it remains a great challenge to localize on-demand release of anticancer drugs in tumor tissues to improve therapeutic effects and minimize the side effects. In this regard, photoresponsive DDSs that employ light as an external stimulus can offer a precise spatiotemporal control of drug release at desired sites of interest. Most photoresponsive DDSs are only responsive to ultraviolet-visible light that shows phototoxicity and/or shallow tissue penetration depth, and thereby their applications are greatly restricted. To address these issues, near-infrared (NIR) photoresponsive DDSs have been developed. In this review, the development of NIR photoresponsive DDSs in last several years for cancer photo-chemotherapy are summarized. They can achieve on-demand release of drugs into tumors of living animals through photothermal, photodynamic, and photoconversion mechanisms, affording obviously amplified therapeutic effects in synergy with phototherapy. Finally, the existing challenges and further perspectives on the development of NIR photoresponsive DDSs and their clinical translation are discussed.
Collapse
Affiliation(s)
- Xiaoying Wang
- Xuhui District Center for Disease Control and Prevention, Shanghai, 200237, China
| | - Zeliang Xuan
- Xuhui District Center for Disease Control and Prevention, Shanghai, 200237, China
| | - Xiaofeng Zhu
- Xuhui District Center for Disease Control and Prevention, Shanghai, 200237, China
| | - Haitao Sun
- Shanghai Institute of Medical Imaging, Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingchao Li
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China.
| |
Collapse
|
191
|
Li Y, Chen M, Yao B, Lu X, Song B, Vasilatos SN, Zhang X, Ren X, Yao C, Bian W, Sun L. Dual pH/ROS-Responsive Nanoplatform with Deep Tumor Penetration and Self-Amplified Drug Release for Enhancing Tumor Chemotherapeutic Efficacy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002188. [PMID: 32627387 DOI: 10.1002/smll.202002188] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/28/2020] [Indexed: 05/13/2023]
Abstract
Poor deep tumor penetration and incomplete intracellular drug release remain challenges for antitumor nanomedicine application in clinical settings. Herein, a nanomedicine (RLPA-NPs) is developed that can achieve prolonged blood circulation, deep tumor penetration, active-targeting of cancer cells, endosome/lysosome escape, and intracellular selectivity self-amplified drug release for effective drug delivery. The RLPA-NPs are constructed by encapsulation of a pH-sensitive polymer octadecylamine-poly(aspartate-1-(3-aminopropyl) imidazole) (OA-P(Asp-API)) and a ROS-generation agent, β-Lapachone (Lap), in micelles assembled by the tumor-penetration peptide internalizing RGD (iRGD)-modified ROS-responsive paclitaxel (PTX)-prodrug. iRGD could promote RLPA-NPs penetration into deep tumor tissue, and specific targeting to cancer cells. After internalization by cancer cells through receptor-mediated endocytosis, OA-P(Asp-API) can rapidly protonate in the endosome's acidic environment, resulting in RLPA-NPs escape from the endosome through the "proton sponge effect". At the same time, the RLPA-NPs micelle disassembles, releasing Lap and PTX-prodrug. Subsequently, the released Lap could generate ROS, consequently amplifying and accelerating PTX release to kill tumor cells. The in vitro and in vivo studies demonstrated that RLPA-NPs can significantly improve the therapeutic effect compared to control groups. Therefore, RLPA-NPs are a promising nanoplatform for overcoming multiple physiological and pathological barriers to enhance drug delivery.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Mie Chen
- Department of General Surgery, Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Nanjing, 211899, China
| | - Bowen Yao
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Xun Lu
- Milken School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Boyang Song
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Shauna N Vasilatos
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiang Zhang
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Shuyang People's Hospital, Suqian, 223600, China
| | - Xiaomei Ren
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Chang Yao
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Weihe Bian
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Lizhu Sun
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Shuyang People's Hospital, Suqian, 223600, China
| |
Collapse
|
192
|
Zhang Y, Xu C, Yang X, Pu K. Photoactivatable Protherapeutic Nanomedicine for Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002661. [PMID: 32667701 DOI: 10.1002/adma.202002661] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/21/2020] [Indexed: 05/24/2023]
Abstract
Therapeutic systems with site-specific pharmaceutical activation hold great promise to enhance therapeutic efficacy while reducing systemic toxicity in cancer therapy. With operational flexibility, noninvasiveness, and high spatiotemporal resolution, photoactivatable nanomedicines have drawn growing attention. Distinct from traditional controlled release systems relying on the difference of biomarker concentrations between disease and healthy tissues, photoactivatable nanomedicines capitalize on the interaction between nanotransducers and light to either trigger photochemical reactions or generate reactive oxygen species (ROS) or heat effect to remotely induce pharmaceutical actions in living subjects. Herein, the recent advances in the development of photoactivatable protherapeutic nanoagents for oncology are summarized. The design strategies and therapeutic applications of these nanoagents are described. Representative examples of each type are discussed in terms of structure, photoactivation mechanism, and preclinical models. Last, potential challenges and perspectives to further develop photoactivatable protherapeutic nanoagents in cancer nanomedicine are discussed.
Collapse
Affiliation(s)
- Yan Zhang
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Cheng Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| | - Xiangliang Yang
- National Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637457, Singapore
| |
Collapse
|
193
|
Xu Y, Lu Q, Sun L, Feng S, Nie Y, Ning X, Lu M. Nanosized Phase-Changeable "Sonocyte" for Promoting Ultrasound Assessment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002950. [PMID: 32697421 DOI: 10.1002/smll.202002950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/16/2020] [Indexed: 05/13/2023]
Abstract
Despite the ability of microbubble contrast agents to improve ultrasound diagnostic performance, their application potential is limited due to low stability, fast clearance, and poor tissue permeation. This study presents a promising nanosized phase-changeable erythrocyte (Sonocyte), composed of liposomal dodecafluoropentane coated with multilayered red blood cell membranes (RBCm), for improving ultrasound assessments. Sonocyte is the first RBCm-functionalized ultrasound contrast agent with uniform nanosized morphology, and exhibits good stability, systemic circulation, target-tissue accumulation, and even ultrasound-responsive phase transition, thereby satisfying the inherent requirement of ultrasound imaging. It is identified that Sonocyte displays similar sensitivity as microbubble SonoVue, a clinical ultrasound contrast agent, for effectively detecting normal parenchyma and hepatic necrosis. Importantly, compared with SonoVue lacking of ability to detect tumors, Sonocyte can identify tumors with high sensitivity and specificity due to superior tumor accumulation and penetration. Therefore, Sonocyte exhibits superior capabilities over SonoVue, endowing with a great clinical application potential.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Qiangbing Lu
- National Laboratory of Solid State Microstructures, Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, China
| | - Lei Sun
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Shujun Feng
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Yuanyuan Nie
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Minghui Lu
- National Laboratory of Solid State Microstructures, Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
194
|
Choi SK. Photoactivation Strategies for Therapeutic Release in Nanodelivery Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences University of Michigan Medical School Ann Arbor MI 48109 USA
- Department of Internal Medicine University of Michigan Medical School Ann Arbor MI 48109 USA
| |
Collapse
|
195
|
Li S, Shan X, Wang Y, Chen Q, Sun J, He Z, Sun B, Luo C. Dimeric prodrug-based nanomedicines for cancer therapy. J Control Release 2020; 326:510-522. [PMID: 32721523 DOI: 10.1016/j.jconrel.2020.07.036] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 01/10/2023]
Abstract
With the rapid development of conjugation chemistry and biomedical nanotechnology, prodrug-based nanosystems (PNS) have emerged as promising drug delivery nanoplatforms. Dimeric prodrug, as an emerging branch of prodrug, has been widely investigated by covalently conjugating two same or different drug molecules. In recent years, great progress has been made in dimeric prodrug-based nanosystems (DPNS) for cancer therapy. Many advantages offered by DPNS have significantly facilitated the delivery efficiency of anticancer drugs, such as high drug loading capacity, favorable pharmacokinetics, tumor stimuli-sensitive drug release and facile combination theranostics. Given the rapid developments in this field, we here outline the latest updates of DPNS in cancer treatment, focusing on dimeric prodrug-encapsulated nanosystems, dimeric prodrug-nanoassemblies and tumor stimuli-responsive DPNS. Moreover, the design principle, advantages and challenges of DPNS for clinical cancer therapy are also highlighted.
Collapse
Affiliation(s)
- Shumeng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinzhu Shan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qin Chen
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
196
|
Zuo W, Chen D, Fan Z, Chen L, Zhu Z, Zhu Q, Zhu X. Design of light/ROS cascade-responsive tumor-recognizing nanotheranostics for spatiotemporally controlled drug release in locoregional photo-chemotherapy. Acta Biomater 2020; 111:327-340. [PMID: 32434075 DOI: 10.1016/j.actbio.2020.04.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/25/2022]
Abstract
Carrier-free nanotheranostics with high drug loading and no carrier-related toxicity are highly promising cancer therapy agents. However, the limited tumor accumulation and poorly controlled drug release of these nanotheranostics continue to be major challenges that restrict clinical applications. In this study, we develop a tumor-recognizing carrier-free nanotheranostic with light/reactive oxygen species (ROS) cascade-responsiveness for spatiotemporally selective photo-chemotherapy. The nanotheranostic is constructed by co-assembly of the indocyanine green (ICG) photosensitizer and the mannose-thioketal-doxorubicin conjugate (MAN-TK-DOX) (abbreviated as IMTD), efficiently preventing premature DOX leakage during blood circulation while reducing nonspecific damage to normal tissues/cells. Once accumulated in tumor tissues, IMTD rapidly diffuses into cancer cells via lectin receptors-mediated endocytosis. Photoacoustic/fluorescence-imaging-guided laser irradiation induces local hyperthermia and ROS generation in tumor cells, thereby promoting apoptosis. Together, the ICG-generated ROS and the endogenous ROS in cancer cells synergistically enhance DOX release, resulting in more efficient chemotherapeutic effects. The in vitro and in vivo results consistently demonstrate that IMTD achieves superior tumor accumulation, highly controllable drug release, and synergetic photo-chemotherapy. Therefore, the co-assembly of an ROS-sensitive targeting ligand-chemodrug conjugate and a photosensitizer could be used to develop spatiotemporally light-activatable nanotheranostics for precision cancer therapy. STATEMENT OF SIGNIFICANCE: Synergistic phototherapy and chemotherapy have been considered as a promising cancer treatment modality to maximize the therapeutic efficacy. Unfortunately, most nanodrugs consisting of chemotherapeutic drug and photosensitizer suffer from suboptimal tumor accumulation and poorly controlled drug release, which results in reduced therapeutic outcome. In this study, Mannose (MAN) was conjugated to the anticancer drug doxorubicin (DOX) by a ROS-sensitive thioketal linker (TK), the obtained amphiphilic MAN-TK-DOX could serve as an ideal self-carrier material to deliver photosensitizer, thus to achieve high-efficient tumor-targeting, spatiotemporal controlled drug release, and superior antitumor effect. We believe that the ROS-sensitive amphiphilic targeting ligand-chemodrug conjugate could be developed as a universal approach for designing tumor-targeted nanodrugs with precisely controlled drug release.
Collapse
Affiliation(s)
- Wenbao Zuo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China
| | - Dengyue Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China
| | - Zhongxiong Fan
- Department of Biomaterials, College of Materials, Research Center of Biomedical Engineering of Xiamen & Key Laboratory of Biomedical Engineering of Fujian Province & Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China
| | - Luping Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China
| | - Zhaoyuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China; School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qixin Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
197
|
Ha SYY, Zhou Y, Fong WP, Ng DKP. Multifunctional Molecular Therapeutic Agent for Targeted and Controlled Dual Chemo- and Photodynamic Therapy. J Med Chem 2020; 63:8512-8523. [DOI: 10.1021/acs.jmedchem.0c00893] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
198
|
Tang Y, Ji Y, Yi C, Cheng D, Wang B, Fu Y, Xu Y, Qian X, Choonara YE, Pillay V, Zhu W, Liu Y, Nie Z. Self-accelerating H 2O 2-responsive Plasmonic Nanovesicles for Synergistic Chemo/starving therapy of Tumors. Am J Cancer Res 2020; 10:8691-8704. [PMID: 32754272 PMCID: PMC7392001 DOI: 10.7150/thno.45392] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Nanoscale vehicles responsive to abnormal variation in tumor environment are promising for use in targeted delivery of therapeutic drugs specifically to tumor sites. Herein, we report the design and fabrication of self-accelerating H2O2-responsive plasmonic gold nanovesicles (GVs) encapsulated with tirapazamine (TPZ) and glucose oxidase (GOx) for synergistic chemo/starving therapy of cancers. Methods: Gold nanoparticles were modified with H2O2-responsive amphiphilic block copolymer PEG45-b-PABE330 by ligand exchange. The TPZ and GOx loaded GVs (TG-GVs) were prepared through the self-assembly of PEG45-b-PABE330 -grafted nanoparticles together with TPZ and GOx by solvent displacement method. Results: In response to H2O2 in tumor, the TG-GVs dissociate to release the payloads that are, otherwise, retained inside the vesicles for days without noticeable leakage. The released GOx enzymes catalyze the oxidation of glucose by oxygen in the tumor tissue to enhance the degree of hypoxia that subsequently triggers the reduction of hypoxia-activated pro-drug TPZ into highly toxic free radicals. The H2O2 generated in the GOx-catalyzed reaction also accelerate the dissociation of vesicles and hence the release rate of the cargoes in tumors. The drug-loaded GVs exhibit superior tumor inhibition efficacy in 4T1 tumor-bearing mice owing to the synergistic effect of chemo/starvation therapy, in addition to their use as contrast agents for computed tomography imaging of tumors. Conclusion: This nanoplatform may find application in managing tumors deeply trapped in viscera or other important tissues that are not compatible with external stimulus (e.g. light).
Collapse
|
199
|
Wang H, Liu Y, He R, Xu D, Zang J, Weeranoppanant N, Dong H, Li Y. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomater Sci 2020; 8:552-568. [PMID: 31769765 DOI: 10.1039/c9bm01392j] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticle capture and elimination by the immune system are great obstacles for drug delivery. Camouflaging nanoparticles with cell membrane represents a promising strategy to communicate and negotiate with the immune system. As a novel class of nanotherapeutics, such biomimetic nanoparticles inherit specific biological functionalities of the source cells (e.g., erythrocytes, immune cells, cancer cells and platelets) in order to evade immune elimination, prolong circulation time, and even target a disease region by virtue of the homing tendency of the cell membrane protein. In this review, we begin with an overview of different cell membranes that can be utilized to create a biointerface on nanoparticles. Subsequently, we elaborate on the state-of-the-art of cell membrane biomimetic nanoparticles for drug delivery. In particular, a summary of data on circulation capacity and targeting efficiency by camouflaged nanoparticles is presented. In addition to cancer therapy, inflammation treatment, as an emerging application of biomimetic nanoparticles, is specifically included. The challenges and outlook of this technology are discussed.
Collapse
Affiliation(s)
- Huaiji Wang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Zhang H, Dong S, Li Z, Feng X, Xu W, Tulinao CMS, Jiang Y, Ding J. Biointerface engineering nanoplatforms for cancer-targeted drug delivery. Asian J Pharm Sci 2020; 15:397-415. [PMID: 32952666 PMCID: PMC7486517 DOI: 10.1016/j.ajps.2019.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
Over the past decade, nanoparticle-based therapeutic modalities have become promising strategies in cancer therapy. Selective delivery of anticancer drugs to the lesion sites is critical for elimination of the tumor and an improved prognosis. Innovative design and advanced biointerface engineering have promoted the development of various nanocarriers for optimized drug delivery. Keeping in mind the biological framework of the tumor microenvironment, biomembrane-camouflaged nanoplatforms have been a research focus, reflecting their superiority in cancer targeting. In this review, we summarize the development of various biomimetic cell membrane-camouflaged nanoplatforms for cancer-targeted drug delivery, which are classified according to the membranes from different cells. The challenges and opportunities of the advanced biointerface engineering drug delivery nanosystems in cancer therapy are discussed.
Collapse
Affiliation(s)
- Huaiyu Zhang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Shujun Dong
- VIP Integrated Department, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | | | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|