1
|
Peng D, Sun S, Zhao M, Zhan L, Wang X. Current Advances in Nanomaterials Affecting Functions and Morphology of Platelets. J Funct Biomater 2024; 15:188. [PMID: 39057309 PMCID: PMC11278457 DOI: 10.3390/jfb15070188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Nanomaterials have been extensively used in the biomedical field due to their unique physical and chemical properties. They promise wide applications in the diagnosis, prevention, and treatment of diseases. Nanodrugs are generally transported to target tissues or organs by coupling targeting molecules or enhanced permeability and retention effect (EPR) passively. As intravenous injection is the most common means of administration of nanomedicine, the transport process inevitably involves the interactions between nanoparticles (NPs) and blood cells. Platelets are known to not only play a critical role in normal coagulation by performing adhesion, aggregation, release, and contraction functions, but also be associated with pathological thrombosis, tumor metastasis, inflammation, and immune reactions, making it necessary to investigate the effects of NPs on platelet function during transport, particularly the way in which their physical and chemical properties determine their interaction with platelets and the underlying mechanisms by which they activate and induce platelet aggregation. However, such data are lacking. This review is intended to summarize the effects of NPs on platelet activation, aggregation, release, and apoptosis, as well as their effects on membrane proteins and morphology in order to shed light on such key issues as how to reduce their adverse reactions in the blood system, which should be taken into consideration in NP engineering.
Collapse
Affiliation(s)
| | | | | | - Linsheng Zhan
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China; (D.P.); (S.S.); (M.Z.)
| | - Xiaohui Wang
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China; (D.P.); (S.S.); (M.Z.)
| |
Collapse
|
2
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
3
|
Wang C, Xu J, Zhang Y, Nie G. Emerging nanotechnological approaches to regulating tumor vasculature for cancer therapy. J Control Release 2023; 362:647-666. [PMID: 37703928 DOI: 10.1016/j.jconrel.2023.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yinlong Zhang
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; School of Nanoscience and Engineering, School of Chemical Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China.
| |
Collapse
|
4
|
Zhu D, Li Y, Zhang Z, Xue Z, Hua Z, Luo X, Zhao T, Lu C, Liu Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021; 19:435. [PMID: 34930293 PMCID: PMC8686559 DOI: 10.1186/s12951-021-01190-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor vessels can provide oxygen and nutrition for solid tumor tissue, create abnormal tumor microenvironment (TME), and play a vital role in the development, immune escape, metastasis and drug resistance of tumor. Tumor vessel-targeting therapy has become an important and promising direction in anti-tumor therapy, with the development of five anti-tumor therapeutic strategies, including vascular disruption, anti-angiogenesis, vascular blockade, vascular normalization and breaking immunosuppressive TME. However, the insufficient drug accumulation and severe side effects of vessel-targeting drugs limit their development in clinical application. Nanotechnology offers an excellent platform with flexible modified surface that can precisely deliver diverse cargoes, optimize efficacy, reduce side effects, and realize the combined therapy. Various nanomedicines (NMs) have been developed to target abnormal tumor vessels and specific TME to achieve more efficient vessel-targeting therapy. The article reviews tumor vascular abnormalities and the resulting abnormal microenvironment, the application of NMs in the tumor vessel-targeting strategies, and how NMs can improve these strategies and achieve multi-strategies combination to maximize anti-tumor effects. ![]()
Collapse
Affiliation(s)
- Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
5
|
Xiong W, Qi L, Si D, Jiang X, Liu Y, Zheng C, Li Y, Shen J, Zhou Z. Effective tumor vessel barrier disruption mediated by perfluoro-N-(4-methylcyclohexyl) piperidine nanoparticles to enhance the efficacy of photodynamic therapy. NANOSCALE 2021; 13:13473-13486. [PMID: 34477752 DOI: 10.1039/d1nr02880d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Currently, limited tumor drug permeation, poor oxygen perfusion and immunosuppressive microenvironments are the most important bottlenecks that significantly reduce the efficacy of photodynamic therapy (PDT). The main cause of these major bottlenecks is the platelet activation maintained abnormal tumor vessel barriers. Thus, platelet inhibition may present a new way to most effectively enhance the efficacy of PDT. However, to the best of our knowledge, few studies have validated the effectiveness of such a way in enhancing the efficacy of PDT both in vivo and in vitro. In this study, perfluoro-N-(4-methylcyclohexyl) piperidine-loaded albumin (PMP@Alb) nanoparticles were discovered, which possess excellent platelet inhibition ability. After PMP@Alb treatment, remarkably enhanced intra-tumoral drug accumulation, oxygen perfusion and T cell infiltration could be observed owing to the disrupted tumor vessel barriers. Besides, the effect of ICG@Lip mediated PDT was significantly amplified by PMP@Alb nanoparticles. It was demonstrated that PMP@Alb could be used as a useful tool to improve the efficacy of existing PDT by disrupting tumor vessel barriers through effective platelet inhibition.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Liu Y, Zhou J, Li Q, Li L, Jia Y, Geng F, Zhou J, Yin T. Tumor microenvironment remodeling-based penetration strategies to amplify nanodrug accessibility to tumor parenchyma. Adv Drug Deliv Rev 2021; 172:80-103. [PMID: 33705874 DOI: 10.1016/j.addr.2021.02.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Remarkable advances in nano delivery systems have provided new hope for tumor prevention, diagnosis and treatment. However, only limited clinical therapeutic effects against solid tumors were achieved. One of the main reasons is the presence of abundant physiological and pathological barriers in vivo that impair tumoral penetration and distribution of the nanodrugs. These barriers are related to the components of tumor microenvironment (TME) including abnormal tumor vasculature, rich composition of the extracellular matrix (ECM), and abundant stroma cells. Herein, we review the advanced strategies of TME remodeling to overcome these biological obstacles against nanodrug delivery. This review aims to offer a perspective guideline for the implementation of promising approaches to facilitate intratumoral permeation of nanodrugs through alleviation of biological barriers. At the same time, we analyze the advantages and disadvantages of the corresponding methods and put forward possible directions for the future researches.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jiyuan Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Qiang Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Lingchao Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yue Jia
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Feiyang Geng
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
7
|
Xu Y, Liu J, Liu Z, Chen G, Li X, Ren H. Damaging Tumor Vessels with an Ultrasound-Triggered NO Release Nanosystem to Enhance Drug Accumulation and T Cells Infiltration. Int J Nanomedicine 2021; 16:2597-2613. [PMID: 33833514 PMCID: PMC8021257 DOI: 10.2147/ijn.s295445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Limited by tumor vascular barriers, restricted intratumoural T cell infiltration and nanoparticles accumulation remain major bottlenecks for anticancer therapy. Platelets are now known to maintain tumor vascular integrity. Therefore, inhibition of tumor-associated platelets may be an effective method to increase T cell infiltration and drug accumulation at tumor sites. Herein, we designed an ultrasound-responsive nitric oxide (NO) release nanosystem, SNO-HSA-PTX, which can release NO in response to ultrasound (US) irradiation, thereby inhibiting platelet function and opening the tumor vascular barrier, promoting drug accumulation and T cell infiltration. METHODS We evaluated the ability of SNO-HSA-PTX to release NO in response to US irradiation. We also tested the effect of SNO-HSA-PTX on platelet function. Plenty of studies including cytotoxicity, pharmacokinetics study, biodistribution, blood perfusion, T cell infiltration, in vivo antitumor efficacy and safety assessment were conducted to investigate the antitumor effect of SNO-HSA-PTX. RESULTS SNO-HSA-PTX with US irradiation inhibited tumor-associated platelets activation and induced openings in the tumor vascular barriers, which promoted the accumulation of SNO-HSA-PTX nanoparticles to the tumor sites. Meanwhile, the damaged vascular barriers allowed oxygen-carrying hemoglobin to infiltrate tumor regions, alleviating hypoxia of the tumor microenvironment. In addition, the intratumoral T cell infiltration was augmented, together with chemotherapy and NO therapy, which greatly inhibited tumor growth. DISCUSSION Our research designed a simple strategy to open the vascular barrier by inhibiting the tumor-associated platelets, which provide new ideas for anti-tumor treatment.
Collapse
Affiliation(s)
- Yan Xu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Jiwei Liu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Zhangya Liu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Guoguang Chen
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Xueming Li
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Hao Ren
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
8
|
Huang D, Sun L, Huang L, Chen Y. Nanodrug Delivery Systems Modulate Tumor Vessels to Increase the Enhanced Permeability and Retention Effect. J Pers Med 2021; 11:124. [PMID: 33672813 PMCID: PMC7917988 DOI: 10.3390/jpm11020124] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The use of nanomedicine for antitumor therapy has been extensively investigated for a long time. Enhanced permeability and retention (EPR) effect-mediated drug delivery is currently regarded as an effective way to bring drugs to tumors, especially macromolecular drugs and drug-loaded pharmaceutical nanocarriers. However, a disordered vessel network, and occluded or embolized tumor blood vessels seriously limit the EPR effect. To augment the EPR effect and improve curative effects, in this review, we focused on the perspective of tumor blood vessels, and analyzed the relationship among abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels, and the EPR effect. In this commentary, nanoparticles including liposomes, micelles, and polymers extravasate through the tumor vasculature, which are based on modulating tumor vessels, to increase the EPR effect, thereby increasing their therapeutic effect.
Collapse
Affiliation(s)
- Dong Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lingna Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Yanzuo Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
9
|
Zhang Y, Han X, Nie G. Responsive and activable nanomedicines for remodeling the tumor microenvironment. Nat Protoc 2021; 16:405-430. [PMID: 33311713 DOI: 10.1038/s41596-020-00421-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/23/2020] [Indexed: 01/19/2023]
Abstract
Here we describe two protocols for the construction of responsive and activable nanomedicines that regulate the tumor microenvironment (TME). The TME is composed of all non-cellular and cellular components surrounding a tumor, including the surrounding blood vessels, immune cells, fibroblasts, signaling molecules, and extracellular matrix and has a crucial role in tumor initiation, growth, and metastasis. Owing to the relatively stable properties of the TME compared to tumor cells, which exhibit frequent genetic mutations and epigenetic changes, therapeutic strategies targeting the TME using multifunctional nanomedicines hold great potential for anti-tumor therapy. By regulating tumor-associated platelets and pancreatic stellate cells (PSCs), the two major players in the TME, we can effectively manipulate the physiological barriers for enhanced drug delivery and significantly improve the tumor penetration and therapeutic efficacy of chemotherapeutics. The preparation and characterization of the multifunctional nanoparticles takes ~10 h for tumor-associated platelet regulation and 16 h for PSC regulation. These nanoformulations can be readily applied to regulate other components in the TME to realize synergistic or additive anti-tumor activity.
Collapse
Affiliation(s)
- Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
- GBA Research Innovation Institute for Nanotechnology, Guangdong, China.
| |
Collapse
|
10
|
Tang S, Zhang F, Gong H, Wei F, Zhuang J, Karshalev E, Esteban-Fernández de Ávila B, Huang C, Zhou Z, Li Z, Yin L, Dong H, Fang RH, Zhang X, Zhang L, Wang J. Enzyme-powered Janus platelet cell robots for active and targeted drug delivery. Sci Robot 2020; 5:5/43/eaba6137. [DOI: 10.1126/scirobotics.aba6137] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/30/2020] [Indexed: 12/14/2022]
Abstract
Transforming natural cells into functional biocompatible robots capable of active movement is expected to enhance the functions of the cells and revolutionize the development of synthetic micromotors. However, present cell-based micromotor systems commonly require the propulsion capabilities of rigid motors, external fields, or harsh conditions, which may compromise biocompatibility and require complex actuation equipment. Here, we report on an endogenous enzyme-powered Janus platelet micromotor (JPL-motor) system prepared by immobilizing urease asymmetrically onto the surface of natural platelet cells. This Janus distribution of urease on platelet cells enables uneven decomposition of urea in biofluids to generate enhanced chemophoretic motion. The cell surface engineering with urease has negligible impact on the functional surface proteins of platelets, and hence, the resulting JPL-motors preserve the intrinsic biofunctionalities of platelets, including effective targeting of cancer cells and bacteria. The efficient propulsion of JPL-motors in the presence of the urea fuel greatly enhances their binding efficiency with these biological targets and improves their therapeutic efficacy when loaded with model anticancer or antibiotic drugs. Overall, asymmetric enzyme immobilization on the platelet surface leads to a biogenic microrobotic system capable of autonomous movement using biological fuel. The ability to impart self-propulsion onto biological cells, such as platelets, and to load these cellular robots with a variety of functional components holds considerable promise for developing multifunctional cell-based micromotors for a variety of biomedical applications.
Collapse
Affiliation(s)
- Songsong Tang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Fangyu Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Hua Gong
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Fanan Wei
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia Zhuang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Emil Karshalev
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Chuying Huang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhidong Zhou
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhengxing Li
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Lu Yin
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Haifeng Dong
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Ronnie H. Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Joseph Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
11
|
Stoiber P, Ekladious I, Zhao Q, Colson YL, Schaus SE, Hansen U, Grinstaff MW. Expansile Nanoparticles Encapsulate Factor Quinolinone Inhibitor 1 and Accumulate in Murine Liver upon Intravenous Administration. Biomacromolecules 2020; 21:1499-1506. [PMID: 32101401 DOI: 10.1021/acs.biomac.0c00064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Expansile nanoparticles (eNPs) are a promising pH-responsive polymeric drug delivery vehicle, as demonstrated in multiple intraperitoneal cancer models. However, previous delivery routes were limited to intraperitoneal injection and to a single agent, paclitaxel. In this study, we preliminarily evaluate the biodistribution and in vivo toxicity of eNPs in mice after intravenous injection. The eNPs localize predominantly to the liver, without detectable acute toxicity in the liver or other key organs. On the basis of these results, we encapsulated FQI1, a promising lead compound for treatment of hepatocellular carcinoma, in eNPs. eNPs are taken up by cancerous and noncancerous human liver cells in vitro, although at different rates. FQI1-loaded eNPs release FQI1 in a pH-dependent manner and limit proliferation equivalently to unencapsulated FQI1 in immortalized hepatocytes in vitro. eNPs are a versatile platform delivery system for therapeutic compounds and have potential utility in the treatment of liver disease.
Collapse
Affiliation(s)
- Patrick Stoiber
- MCBB Graduate Program and Department of Biology, Boston University, Boston, Massachusetts 02215, United States
| | - Iriny Ekladious
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Qing Zhao
- Department of Pathology and Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts 02118, United States
| | - Yolonda L Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Scott E Schaus
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States.,Center for Molecular Discovery, Boston University, Boston, Massachusetts 02215, United States
| | - Ulla Hansen
- MCBB Graduate Program and Department of Biology, Boston University, Boston, Massachusetts 02215, United States
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States.,Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
12
|
Perfluorocarbon nanoparticle-mediated platelet inhibition promotes intratumoral infiltration of T cells and boosts immunotherapy. Proc Natl Acad Sci U S A 2019; 116:11972-11977. [PMID: 31142648 DOI: 10.1073/pnas.1901987116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immunotherapy can stimulate and enhance the ability of the immune system to recognize, arrest, and eliminate tumor cells. Immune checkpoint therapies (e.g., PD-1/PD-L1) have shown an unprecedented and durable clinical response rate in patients among various cancer types. However, a large fraction of patients still does not respond to these checkpoint inhibitors. The main cause of this phenomenon is the limited T-cell infiltration in tumors. Therefore, additional strategies to enhance T-cell trafficking into tumors are urgently needed to improve patients' immune responses. In this study, we screened an array of perfluorocarbon compounds, reporting that albumin-based perfluorotributylamine nanoparticles (PFTBA@Alb) can effectively increase the permeability of tumor blood vessels, and no distinct side effects were found on normal blood vessels. After i.v. administration of PFTBA@Alb, the number of tumor-infiltrating CD8+ and CD4+ T cells showed an obvious rising trend. More important, a striking tumor inhibition rate, reaching nearly 90%, was observed when combining PFTBA@Alb with anti-PD-L1 antibody. These findings suggest that PFTBA@Alb can be regarded as an enhancer for anti-PD-L1 immunotherapy.
Collapse
|
13
|
Ye H, Wang K, Wang M, Liu R, Song H, Li N, Lu Q, Zhang W, Du Y, Yang W, Zhong L, Wang Y, Yu B, Wang H, Kan Q, Zhang H, Wang Y, He Z, Sun J. Bioinspired nanoplatelets for chemo-photothermal therapy of breast cancer metastasis inhibition. Biomaterials 2019; 206:1-12. [PMID: 30921730 DOI: 10.1016/j.biomaterials.2019.03.024] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 11/16/2022]
Abstract
Breast cancer is associated with high mortality due to tumor metastasis. The anti-metastasis efficacy of photochemotherapy is strictly limited by poor targeting capability with respect to circulating tumor cells (CTCs) in blood and lymph. Herein, we decorate the platelet membrane (PM) on a surface of nanoparticles (NPs), referred to as nanoplatelets. A chemotherapeutic drug, doxorubicin (DOX), and an FDA-approved photothermal agent, indocyanine green (ICG), are co-encapsulated into the biomimetic nanoplatelets. Nanoplatelets possess immune surveillance-escaping capability and specifically capture and clear CTCs in both blood and lymphatic circulations via high-affinity interactions between the P-Selectin of PM and CD44 receptors of tumor cells. PM-coated NPs show greater cellular uptake in MDA-MB-231 breast cancer cells and further elicit higher cytotoxicity to tumor cells relative to uncoated NPs. In vivo, we disclose that the multifunctional nanoplatelets not only completely ablate the primary tumor but also inhibit breast cancer metastasis with high efficiency in the three established xenograft or orthotopic breast tumor-bearing mice models. We conclude that such biomimetic nanoplatelets represent a promising strategy of coating a surface of nanoparticles with platelet membrane to actively capture and destroy CTCs in blood and lymph in breast cancer anti-metastasis therapy.
Collapse
Affiliation(s)
- Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Menglin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Rongzheng Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Hang Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Na Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Wenjuan Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yuqian Du
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Wenqian Yang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Lu Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yu Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Bohong Yu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Hong Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Qiming Kan
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Yongjun Wang
- 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.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
14
|
Brighi C, Puttick S, Rose S, Whittaker AK. The potential for remodelling the tumour vasculature in glioblastoma. Adv Drug Deliv Rev 2018; 136-137:49-61. [PMID: 30308226 DOI: 10.1016/j.addr.2018.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/19/2022]
Abstract
Despite significant improvements in the clinical management of glioblastoma, poor delivery of systemic therapies to the entire population of tumour cells remains one of the biggest challenges in the achievement of more effective treatments. On the one hand, the abnormal and dysfunctional tumour vascular network largely limits blood perfusion, resulting in an inhomogeneous delivery of drugs to the tumour. On the other hand, the presence of an intact blood-brain barrier (BBB) in certain regions of the tumour prevents chemotherapeutic drugs from permeating through the tumour vessels and reaching the diseased cells. In this review we analyse in detail the implications of the presence of a dysfunctional vascular network and the impenetrable BBB on drug transport. We discuss advantages and limitations of the currently available strategies for remodelling the tumour vasculature aiming to ameliorate the above mentioned limitations. Finally we review research methods for visualising vascular dysfunction and highlight the power of DCE- and DSC-MRI imaging to assess changes in blood perfusion and BBB permeability.
Collapse
|
15
|
Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018. [PMID: 29519806 DOI: 10.1182/blood-2017-05-743187] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Platelets have long been recognized as key players in hemostasis and thrombosis; however, growing evidence suggests that they are also significantly involved in cancer, the second leading cause of mortality worldwide. Preclinical and clinical studies showed that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between platelets and cancer cells. For example, cancer changes platelet behavior by directly inducing tumor-platelet aggregates, triggering platelet granule and extracellular vesicle release, altering platelet phenotype and platelet RNA profiles, and enhancing thrombopoiesis. Reciprocally, platelets reinforce tumor growth with proliferation signals, antiapoptotic effect, and angiogenic factors. Platelets also activate tumor invasion and sustain metastasis via inducing an invasive epithelial-mesenchymal transition phenotype of tumor cells, promoting tumor survival in circulation, tumor arrest at the endothelium, and extravasation. Furthermore, platelets assist tumors in evading immune destruction. Hence, cancer cells and platelets maintain a complex, bidirectional communication. Recently, aspirin (acetylsalicylic acid) has been recognized as a promising cancer-preventive agent. It is recommended at daily low dose by the US Preventive Services Task Force for primary prevention of colorectal cancer. The exact mechanisms of action of aspirin in chemoprevention are not very clear, but evidence has emerged that suggests a platelet-mediated effect. In this article, we will introduce how cancer changes platelets to be more cancer-friendly and highlight advances in the modes of action for aspirin in cancer prevention. We also discuss the opportunities, challenges, and opposing viewpoints on applying aspirin and other antiplatelet agents for cancer prevention and treatment.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - George M Yousef
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, and
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada; and
- Department of Medicine and
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Boulaftali Y, Mawhin M, Jandrot‐Perrus M, Ho‐Tin‐Noé B. Glycoprotein VI in securing vascular integrity in inflamed vessels. Res Pract Thromb Haemost 2018; 2:228-239. [PMID: 30046725 PMCID: PMC5974920 DOI: 10.1002/rth2.12092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Glycoprotein VI (GPVI), the main platelet receptor for collagen, has been shown to play a central role in various models of thrombosis, and to be a minor actor of hemostasis at sites of trauma. These observations have made of GPVI a novel target for antithrombotic therapy, as its inhibition would ideally combine efficacy with safety. Nevertheless, recent studies have indicated that GPVI could play an important role in preventing bleeding caused by neutrophils in the inflamed skin and lungs. Remarkably, there is evidence that the GPVI-dependent hemostatic function of platelets at the acute phase of inflammation in these organs does not involve aggregation. From a therapeutic perspective, the vasculoprotective action of GPVI in inflammation suggests that blocking of GPVI might bear some risks of bleeding at sites of neutrophil infiltration. In this review, we summarize recent findings on GPVI functions in inflammation and discuss their possible clinical implications and applications.
Collapse
Affiliation(s)
- Yacine Boulaftali
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Marie‐Anne Mawhin
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Martine Jandrot‐Perrus
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Benoît Ho‐Tin‐Noé
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| |
Collapse
|
17
|
Platelets and vascular integrity: how platelets prevent bleeding in inflammation. Blood 2017; 131:277-288. [PMID: 29191915 DOI: 10.1182/blood-2017-06-742676] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023] Open
Abstract
Platelets play a central role in primary hemostasis by forming aggregates that plug holes in injured vessels. Half a century ago, detailed studies of the microvasculature by electron microscopy revealed that under inflammatory conditions that do not induce major disruption to vascular structure, individual platelets are mobilized to the vessel wall, where they interact with leukocytes and appear to seal gaps that arise between endothelial cells. Recent developments in genetic engineering and intravital microscopy have allowed further molecular and temporal characterization of these events. Surprisingly, it turns out that platelets support the recruitment of leukocytes to sites of inflammation. In parallel, however, they exercise their hemostatic function by securing the integrity of inflamed blood vessels to prevent bleeding from sites of leukocyte infiltration. It thus appears that platelets not only serve in concert as building blocks of the hemostatic plug but also act individually as gatekeepers of the vascular wall to help preserve vascular integrity while coordinating host defense. Variants of this recently appreciated hemostatic function of platelets that we refer to as "inflammation-associated hemostasis" are engaged in different contexts in which the endothelium is challenged or dysfunctional. Although the distinguishing characteristics of these variants and the underlying mechanisms of inflammation-associated hemostasis remain to be fully elucidated, they can differ notably from those supporting thrombosis, thus presenting therapeutic opportunities.
Collapse
|
18
|
Li S, Zhang Y, Wang J, Zhao Y, Ji T, Zhao X, Ding Y, Zhao X, Zhao R, Li F, Yang X, Liu S, Liu Z, Lai J, Whittaker AK, Anderson GJ, Wei J, Nie G. Nanoparticle-mediated local depletion of tumour-associated platelets disrupts vascular barriers and augments drug accumulation in tumours. Nat Biomed Eng 2017; 1:667-679. [PMID: 31015598 DOI: 10.1038/s41551-017-0115-8] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 06/08/2017] [Indexed: 11/09/2022]
Abstract
Limited intratumoural perfusion and nanoparticle retention remain major bottlenecks for the delivery of nanoparticle therapeutics into tumours. Here, we show that polymer-lipid-peptide nanoparticles delivering the antiplatelet antibody R300 and the chemotherapeutic agent doxorubicin can locally deplete tumour-associated platelets, thereby enhancing vascular permeability and augmenting the accumulation of the nanoparticles in tumours. R300 is specifically released in the tumour on cleavage of the lipid-peptide shell of the nanoparticles by matrix metalloprotease 2, which is commonly overexpressed in tumour vascular endothelia and stroma, thus facilitating vascular breaches that enhance tumour permeability. We also show that this strategy leads to substantial tumour regression and metastasis inhibition in mice.
Collapse
Affiliation(s)
- Suping Li
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yinlong Zhang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Jing Wang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ying Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Tianjiao Ji
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiao Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yanping Ding
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiaozheng Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ruifang Zhao
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Feng Li
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xiao Yang
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Shaoli Liu
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jianhao Lai
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Andrew K Whittaker
- Australian Institute for Bioengineering and Nanotechnology, Centre for Magnetic Resonance, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gregory J Anderson
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Jingyan Wei
- College of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Guangjun Nie
- Chinese Academy of Sciences Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Chinese Academy of Sciences Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
19
|
Baigger A, Blasczyk R, Figueiredo C. Towards the Manufacture of Megakaryocytes and Platelets for Clinical Application. Transfus Med Hemother 2017. [PMID: 28626367 DOI: 10.1159/000477261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Platelet transfusions are used in standard clinical practice to prevent hemorrhage in patients suffering from thrombocytopenia or platelet dysfunctions. Recently, a constant rise on the demand of platelets for transfusion has been registered. This may be associated with several factors including demographic changes, population aging as well as incidence and prevalence of hematological diseases. In addition, platelet-regenerative properties have been started to be exploited in different areas such as tissue remodeling and anti-cancer therapies. These new applications are also expected to increase the future demand on platelets. Thus, in vitro generated platelets may constitute a highly desirable alternative to meet the rising demand on platelets. Several factors have been considered in the road trip of producing in vitro megakaryocytes and platelets for clinical application. From selection of the cell source, differentiation protocols and culture conditions to the design of optimal bioreactors, several strategies have been proposed to maximize production yields while preserving functionality. This review summarizes new advances in megakaryocyte and platelet differentiation and their production upscaling.
Collapse
Affiliation(s)
- Anja Baigger
- Institute for Transfusion Medicine, Hanover Medical School, Hanover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hanover Medical School, Hanover, Germany
| | | |
Collapse
|
20
|
Ayesa U, Gray BD, Pak KY, Chong PLG. Liposomes Containing Lipid-Soluble Zn(II)-Bis-dipicolylamine Derivatives Show Potential To Be Targeted to Phosphatidylserine on the Surface of Cancer Cells. Mol Pharm 2016; 14:147-156. [PMID: 28043132 DOI: 10.1021/acs.molpharmaceut.6b00760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Here we used a lipid-soluble Zn(II)-bis-dipicolylamine derivative as a membrane component to develop liposomal carriers that have potential to be targeted to phosphatidylserine (PS) rich surfaces on cancer cells and to preferentially kill cancer cells without using anticancer drugs. This DPA derivative (abbreviated as DPA-Cy3[22,22]) contains the fluorophore cyanine 3 (Cy3) and two 22-carbon chains that can be anchored into liposomal membrane bilayers. DPA-Cy3[22,22]/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) unilamellar vesicles (∼150 nm) showed selective binding to PS-containing liposomes as demonstrated by anion exchange chromatography. This binding does not result in vesicle fusion or aggregation. Flow cytometry showed that DPA-Cy3[22,22]/POPC liposomes have preferential binding to MCF-7 breast cancer cells over MCF-12A noncancer cells due to 3-7 times more PS exposures on MCF-7. The extent of liposome binding with MCF-7 cells was increased by two times after cells were pretreated with the apoptotic inducer camptothecin, which increased PS exposure to the cell surface. Moreover, our flow cytometry data also suggest that local cell membrane perturbations may occur upon liposome binding and internalization. This implies that DPA-Cy3[22,22]/POPC liposomes alone may have a PS-dependent cytotoxic effect. This assertion was supported by the cell proliferation assay, which showed that 9.1 mol % DPA-Cy3[22,22]/POPC liposomes exert cytotoxicity on MCF-7 cells 3.5 times higher than that on MCF-12A cells. These results indicate that DPA-Cy3[22,22]-containing liposomes hold great promise as efficient nano drug carriers.
Collapse
Affiliation(s)
- Umme Ayesa
- Department of Medical Genetics and Molecular Biochemistry, The Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania 19140, United States
| | - Brian D Gray
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Parkson Lee-Gau Chong
- Department of Medical Genetics and Molecular Biochemistry, The Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
21
|
D'Alessandro R, Messa C, Refolo MG, Carr BI. Modulation of sensitivity and resistance to multikinase inhibitors by microenvironmental platelet factors in HCC. Expert Opin Pharmacother 2015; 16:2773-80. [PMID: 26479083 DOI: 10.1517/14656566.2015.1101065] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Response of a tumor to chemotherapy or multikinase inhibitor therapy has been traditionally thought to be a reflection of the sum of the characteristics of both the drug and of the tumor cell resistance mechanisms. More recently, there has been a growing awareness of the role of non-tumor factors-both cellular and humoral-in the tumor microenvironment that can increase or decrease the tumor cellular responses to the therapy. This article focuses on platelet factors in clinical HCC and experimental evidence that they provide growth stimulants that can antagonize the growth inhibitory effects of therapy. AREAS COVERED Review of the mechanisms of multikinase cancer growth inhibitors and of the role of platelets in providing growth factors that can antagonize their effects. EXPERT OPINION These new ideas and data show that the response of a tumor to multikinase inhibitors or chemotherapy may be strongly influenced by microenvironmental factors. Conversely, antagonists to these environmental factors, such as EGFR inhibitors and IGF1-R inhibitors, might be expected to augment the anti-tumor effect of both chemotherapy and multikinase inhibitors.
Collapse
Affiliation(s)
- Rosalba D'Alessandro
- a Laboratory of Cellular Biology, National Institute for Digestive Diseases , IRCCS "Saverio de Bellis" , Via Turi 27, Castellana Grotte , BA 70013 , Italy
| | - Caterina Messa
- a Laboratory of Cellular Biology, National Institute for Digestive Diseases , IRCCS "Saverio de Bellis" , Via Turi 27, Castellana Grotte , BA 70013 , Italy
| | - Maria Grazia Refolo
- a Laboratory of Cellular Biology, National Institute for Digestive Diseases , IRCCS "Saverio de Bellis" , Via Turi 27, Castellana Grotte , BA 70013 , Italy
| | - Brian I Carr
- a Laboratory of Cellular Biology, National Institute for Digestive Diseases , IRCCS "Saverio de Bellis" , Via Turi 27, Castellana Grotte , BA 70013 , Italy.,b Visiting Professor, Program for Targeted Experimental Therapeutics, Izmir Biomedicine and Genome Center , Dokuz Eylul University , Izmir , Turkey
| |
Collapse
|
22
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
23
|
D'Alessandro R, Refolo MG, Lippolis C, Giannuzzi G, Carella N, Messa C, Cavallini A, Carr BI. Antagonism of sorafenib and regorafenib actions by platelet factors in hepatocellular carcinoma cell lines. BMC Cancer 2014; 14:351. [PMID: 24885890 PMCID: PMC4038078 DOI: 10.1186/1471-2407-14-351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/19/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Platelets are frequently altered in hepatocellular carcinoma (HCC) patients. Platelet lysates (hPL) can enhance HCC cell growth and decrease apoptosis. The aims were to evaluate whether hPL can modulate the actions of sorafenib or regorafenib, two clinical HCC multikinase antagonists. METHODS Several human HCC cell lines were grown in the presence and absence of sorafenib or regorafenib, with or without hPL. Growth was measured by MTT assay, apoptosis was assessed by Annexin V and by western blot, and autophagy and MAPK growth signaling were also measured by western blot, and migration and invasion were measured by standard in vitro assays. RESULTS Both sorafenib and regorafenib-mediated inhibition of cell growth, migration and invasion were all antagonized by hPL. Drug-mediated apoptosis and decrease in phospho-ERK levels were both blocked by hPL, which also increased anti-apoptotic phospho-STAT, Bax and Bcl-xL levels. Preliminary data, obtained with epidermal growth factor (EGF) and insulin-like growth factor-I (IGF-I), included in hPL, revealed that these factors were able to antagonized sorafenib in a proliferation assay, in particular when used in combination. CONCLUSIONS Platelet factors can antagonize sorafenib or regorafenib-mediated growth inhibition and apoptosis in HCC cells. The modulation of platelet activity or numbers has the potential to enhance multikinase drug actions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Brian I Carr
- Laboratory of Biochemistry, National Institute for Digestive Diseases, IRCCS "Saverio de Bellis", Via Turi 27, 70013, Castellana Grotte, BA, Italy.
| |
Collapse
|
24
|
Noble GT, Stefanick JF, Ashley JD, Kiziltepe T, Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol 2013; 32:32-45. [PMID: 24210498 DOI: 10.1016/j.tibtech.2013.09.007] [Citation(s) in RCA: 353] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 12/18/2022]
Abstract
Nanomedicine, particularly liposomal drug delivery, has expanded considerably over the past few decades, and several liposomal drugs are already providing improved clinical outcomes. Liposomes have now progressed beyond simple, inert drug carriers and can be designed to be highly responsive in vivo, with active targeting, increased stealth, and controlled drug-release properties. Ligand-targeted liposomes (LTLs) have the potential to revolutionize the treatment of cancer. However, these highly engineered liposomes generate new problems, such as accelerated clearance from circulation, compromised targeting owing to non-specific serum protein binding, and hindered tumor penetration. This article highlights recent challenges facing LTL strategies and describes the advanced design elements used to circumvent them.
Collapse
Affiliation(s)
- Gavin T Noble
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jared F Stefanick
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jonathan D Ashley
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Tanyel Kiziltepe
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Basar Bilgicer
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Rare & Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
25
|
Sengupta S, Kulkarni A. Design principles for clinical efficacy of cancer nanomedicine: a look into the basics. ACS NANO 2013; 7:2878-82. [PMID: 23607425 PMCID: PMC3876731 DOI: 10.1021/nn4015399] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
With recent advances in cancer nanomedicine, there is an increasing expectation for clinical translation. However, what are the parameters of a nanomedicine that will define clinical success, which will be measured by increased efficacy and not just ease of delivery or reduction in toxicity? In this Perspective, we build on a fundamental study by Stefanick et al. on the significance of the design principles in the engineering of a nanomedicine, such as peptide-PEG-linker length and ligand density in cellular uptake of liposomal nanoparticles. We address additional design parameters that can potentially facilitate clinical translation as well as how emerging insights into tumor biology will inspire next-generation cancer nanomedicines.
Collapse
Affiliation(s)
- Shiladitya Sengupta
- Laboratory of Nanomedicine, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard-MIT Division of Health Science Technology, Harvard Medical School, Dana Farber Cancer Center, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02139, United States.
| | | |
Collapse
|