1
|
Wang M, Liu H, Huang J, Cai T, Xu ZP, Zhang L. Advancing cancer gene therapy: the emerging role of nanoparticle delivery systems. J Nanobiotechnology 2025; 23:362. [PMID: 40394591 PMCID: PMC12090605 DOI: 10.1186/s12951-025-03433-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
Gene therapy holds immense potential due to its ability to precisely target oncogenes, making it a promising strategy for cancer treatment. Advances in genetic science and bioinformatics have expanded the applications of gene delivery technologies beyond detection and diagnosis to potential therapeutic interventions. However, traditional gene therapy faces significant challenges, including limited therapeutic efficacy and the rapid degradation of genetic materials in vivo. To address these limitations, multifunctional nanoparticles have been engineered to encapsulate and protect genetic materials, enhancing their stability and therapeutic effectiveness. Nanoparticles are being extensively explored for their ability to deliver various genetic payloads-including plasmid DNA, messenger RNA, and small interfering RNA-directly to cancer cells. This review highlights key gene modulation strategies such as RNA interference, gene editing systems, and chimeric antigen receptor (CAR) technologies, alongside a diverse array of nanoscale delivery systems composed of polymers, lipids, and inorganic materials. These nanoparticle-based delivery platforms aim to improve targeted transport of genetic material into cancer cells, ultimately enhancing the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Maoze Wang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Huina Liu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
| | - Jinling Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
| | - Zhi Ping Xu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus C, DK-8000, Denmark.
| |
Collapse
|
2
|
Suresh A, Suresh D, Li Z, Sansalone J, Aluru N, Upendran A, Kannan R. Self-Assembled Multilayered Concentric Supraparticle Architecture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502055. [PMID: 40285599 DOI: 10.1002/adma.202502055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Supraparticles (SPs) with unique properties are emerging as versatile platforms for applications in catalysis, photonics, and medicine. However, the synthesis of novel SPs with complex internal structures remains a challenge. Self-Assembled Multilayered Supraparticles (SAMS) presented here are composed of concentric lamellar spherical structures made from metallic nanoparticles, formed from a synergistic three-way interaction phenomenon between gold nanoparticles, lipidoid, and gelatin, exhibiting interlayer spacing of 3.5 ± 0.2 nm within a self-limited 156.8 ± 56.6 nm diameter. The formation is critically influenced by both physical (including nanoparticle size, lipidoid chain length) and chemical factors (including elemental composition, nanoparticle cap, and organic material), which collectively modulate the surface chemistry and hydrophobicity, affecting interparticle interactions. SAMS can efficiently deliver labile payloads such as siRNA, achieving dose-dependent silencing in vivo, while also showing potential for complex payloads such as mRNA. This work not only advances the field of SP design by introducing a new structure and interaction phenomenon but also demonstrates its potential in nanomedicine.
Collapse
Affiliation(s)
- Agasthya Suresh
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO, 65211, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, 65212, USA
| | - Dhananjay Suresh
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
| | - Zhaohui Li
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
| | - John Sansalone
- Department of Mechanical Engineering, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Narayana Aluru
- Department of Mechanical Engineering, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Anandhi Upendran
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, 65212, USA
| | - Raghuraman Kannan
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO, 65211, USA
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, 65212, USA
| |
Collapse
|
3
|
Kovtareva S, Kusepova L, Tazhkenova G, Mashan T, Bazarbaeva K, Kopishev E. Surface Modification of Mesoporous Silica Nanoparticles for Application in Targeted Delivery Systems of Antitumour Drugs. Polymers (Basel) 2024; 16:1105. [PMID: 38675024 PMCID: PMC11054758 DOI: 10.3390/polym16081105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The problem of tumour therapy has attracted the attention of many researchers for many decades. One of the promising strategies for the development of new dosage forms to improve oncology treatment efficacy and minimise side effects is the development of nanoparticle-based targeted transport systems for anticancer drugs. Among inorganic nanoparticles, mesoporous silica deserves special attention due to its outstanding surface properties and drug-loading capability. This review analyses the various factors affecting the cytotoxicity, cellular uptake, and biocompatibility of mesoporous silica nanoparticles (MSNs), constituting a key aspect in the development of safe and effective drug delivery systems. Special attention is paid to technological approaches to chemically modifying MSNs to alter their surface properties. The stimuli that regulate drug release from nanoparticles are also discussed, contributing to the effective control of the delivery process in the body. The findings emphasise the importance of modifying MSNs with different surface functional groups, bio-recognisable molecules, and polymers for their potential use in anticancer drug delivery systems.
Collapse
Affiliation(s)
| | | | | | | | | | - Eldar Kopishev
- Department of Chemistry, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Astana 010000, Kazakhstan; (S.K.); (L.K.); (G.T.); (T.M.); (K.B.)
| |
Collapse
|
4
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
5
|
Neary MT, Mulder LM, Kowalski PS, MacLoughlin R, Crean AM, Ryan KB. Nebulised delivery of RNA formulations to the lungs: From aerosol to cytosol. J Control Release 2024; 366:812-833. [PMID: 38101753 DOI: 10.1016/j.jconrel.2023.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
In the past decade RNA-based therapies such as small interfering RNA (siRNA) and messenger RNA (mRNA) have emerged as new and ground-breaking therapeutic agents for the treatment and prevention of many conditions from viral infection to cancer. Most clinically approved RNA therapies are parenterally administered which impacts patient compliance and adds to healthcare costs. Pulmonary administration via inhalation is a non-invasive means to deliver RNA and offers an attractive alternative to injection. Nebulisation is a particularly appealing method due to the capacity to deliver large RNA doses during tidal breathing. In this review, we discuss the unique physiological barriers presented by the lung to efficient nebulised RNA delivery and approaches adopted to circumvent this problem. Additionally, the different types of nebulisers are evaluated from the perspective of their suitability for RNA delivery. Furthermore, we discuss recent preclinical studies involving nebulisation of RNA and analysis in in vitro and in vivo settings. Several studies have also demonstrated the importance of an effective delivery vector in RNA nebulisation therefore we assess the variety of lipid, polymeric and hybrid-based delivery systems utilised to date. We also consider the outlook for nebulised RNA medicinal products and the hurdles which must be overcome for successful clinical translation. In summary, nebulised RNA delivery has demonstrated promising potential for the treatment of several lung-related conditions such as asthma, COPD and cystic fibrosis, to which the mode of delivery is of crucial importance for clinical success.
Collapse
Affiliation(s)
- Michael T Neary
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | | | - Piotr S Kowalski
- School of Pharmacy, University College Cork, Ireland; APC Microbiome, University College Cork, Cork, Ireland
| | | | - Abina M Crean
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | - Katie B Ryan
- SSPC, The SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland.
| |
Collapse
|
6
|
Ghodasara A, Raza A, Wolfram J, Salomon C, Popat A. Clinical Translation of Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2301010. [PMID: 37421185 DOI: 10.1002/adhm.202301010] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Indexed: 07/10/2023]
Abstract
Extracellular vesicles (EVs) occur in a variety of bodily fluids and have gained recent attraction as natural materials due to their bioactive surfaces, internal cargo, and role in intercellular communication. EVs contain various biomolecules, including surface and cytoplasmic proteins; and nucleic acids that are often representative of the originating cells. EVs can transfer content to other cells, a process that is thought to be important for several biological processes, including immune responses, oncogenesis, and angiogenesis. An increased understanding of the underlying mechanisms of EV biogenesis, composition, and function has led to an exponential increase in preclinical and clinical assessment of EVs for biomedical applications, such as diagnostics and drug delivery. Bacterium-derived EV vaccines have been in clinical use for decades and a few EV-based diagnostic assays regulated under Clinical Laboratory Improvement Amendments have been approved for use in single laboratories. Though, EV-based products are yet to receive widespread clinical approval from national regulatory agencies such as the United States Food and Drug Administration (USFDA) and European Medicine Agency (EMA), many are in late-stage clinical trials. This perspective sheds light on the unique characteristics of EVs, highlighting current clinical trends, emerging applications, challenges and future perspectives of EVs in clinical use.
Collapse
Affiliation(s)
- Aayushi Ghodasara
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Aun Raza
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- The School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- Department of Research, Postgraduate and Further Education (DIPEC), Falcuty of Health Sciences, University of Alba, Santiago, 8320000, Chile
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
| |
Collapse
|
7
|
Amruta A, Iannotta D, Cheetham SW, Lammers T, Wolfram J. Vasculature organotropism in drug delivery. Adv Drug Deliv Rev 2023; 201:115054. [PMID: 37591370 PMCID: PMC10693934 DOI: 10.1016/j.addr.2023.115054] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/22/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Over the past decades, there has been an exponential increase in the development of preclinical and clinical nanodelivery systems, and recently, an accelerating demand to deliver RNA and protein-based therapeutics. Organ-specific vasculature provides a promising intermediary for site-specific delivery of nanoparticles and extracellular vesicles to interstitial cells. Endothelial cells express organ-specific surface marker repertoires that can be used for targeted delivery. This article highlights organ-specific vasculature properties, nanodelivery strategies that exploit vasculature organotropism, and overlooked challenges and opportunities in targeting and simultaneously overcoming the endothelial barrier. Impediments in the clinical translation of vasculature organotropism in drug delivery are also discussed.
Collapse
Affiliation(s)
- A Amruta
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dalila Iannotta
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Seth W Cheetham
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO-ABCD), 52074 Aachen, Germany
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
9
|
Advances in the Application of Nanomaterials to the Treatment of Melanoma. Pharmaceutics 2022; 14:pharmaceutics14102090. [PMID: 36297527 PMCID: PMC9610396 DOI: 10.3390/pharmaceutics14102090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022] Open
Abstract
Melanoma can be divided into cutaneous melanoma, uveal melanoma, mucosal melanoma, etc. It is a very aggressive tumor that is prone to metastasis. Patients with metastatic melanoma have a poor prognosis and shorter survival. Although current melanoma treatments have been dramatically improved, there are still many problems such as systemic toxicity and the off-target effects of drugs. The use of nanoparticles may overcome some inadequacies of current melanoma treatments. In this review, we summarize the limitations of current therapies for cutaneous melanoma, uveal melanoma, and mucosal melanoma, as well as the adjunct role of nanoparticles in different treatment modalities. We suggest that nanomaterials may have an effective intervention in melanoma treatment in the future.
Collapse
|
10
|
Wang J, Lv F, Sun T, Zhao S, Chen H, Liu Y, Liu Z. Sorafenib Nanomicelles Effectively Shrink Tumors by Vaginal Administration for Preoperative Chemotherapy of Cervical Cancer. NANOMATERIALS 2021; 11:nano11123271. [PMID: 34947619 PMCID: PMC8705954 DOI: 10.3390/nano11123271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
To investigate the potential of sorafenib (SF) in preoperative chemotherapy for cervical cancer to reduce tumor volume, sorafenib micelles (SF micelles) with good stability and high drug loading were designed. SF micelles were prepared by film hydration followed by the ultrasonic method. The results showed that the SF micelles were spherical with an average particle size of 67.18 ± 0.66 nm (PDI 0.17 ± 0.01), a considerable drug loading of 15.9 ± 0.46% (w/w%) and satisfactory stability in buffers containing plasma or not for at least 2 days. In vitro release showed that SF was gradually released from SF micelles and almost completely released on the third day. The results of in vitro cellular intake, cytotoxicity and proliferation of cervical cancer cell TC-1 showed that SF micelles were superior to sorafenib (Free SF). For intravaginal administration, SF micelles were dispersed in HPMC (SF micelles/HPMC), showed good viscosity sustained-release profiles in vitro and exhibited extended residence in intravaginal in vivo. Compared with SF micelles dispersed in N.S. (SF micelles/N.S.), SF micelles/HPMC significantly reduced tumor size with a tumor weight inhibition rate of 73%. The results suggested that SF micelles had good potential for preoperative tumor shrinkage and improving the quality life of patients.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China;
| | - Fengmei Lv
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Tao Sun
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Shoujin Zhao
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Haini Chen
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China;
- Correspondence: (Y.L.); (Z.L.)
| | - Zhepeng Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (F.L.); (T.S.); (S.Z.); (H.C.)
- Correspondence: (Y.L.); (Z.L.)
| |
Collapse
|
11
|
Iannotta D, Yang M, Celia C, Di Marzio L, Wolfram J. Extracellular vesicle therapeutics from plasma and adipose tissue. NANO TODAY 2021; 39:101159. [PMID: 33968157 PMCID: PMC8104307 DOI: 10.1016/j.nantod.2021.101159] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Extracellular vesicles (EVs) are cell-released lipid-bilayer nanoparticles that contain biologically active cargo involved in physiological and pathological intercellular communication. In recent years, the therapeutic potential of EVs has been explored in various disease models. In particular, mesenchymal stromal cell-derived EVs have been shown to exert anti-inflammatory, anti-oxidant, anti-apoptotic, and pro-angiogenic properties in cardiovascular, metabolic and orthopedic conditions. However, a major drawback of EV-based therapeutics is scale-up issues due to extensive cell culture requirements and inefficient isolation protocols. An emerging alternative approach to time-consuming and costly cell culture expansion is to obtain therapeutic EVs directly from the body, for example, from plasma and adipose tissue. This review discusses isolation methods and therapeutic applications of plasma and adipose tissue-derived EVs, highlighting advantages and disadvantages compared to cell culture-derived ones.
Collapse
Affiliation(s)
- Dalila Iannotta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Man Yang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Christian Celia
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti – Pescara “G d’Annunzio”, Chieti, Italy
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston TX, USA
| |
Collapse
|
12
|
Zhang LY, Yang X, Wang SB, Chen H, Pan HY, Hu ZM. Membrane Derived Vesicles as Biomimetic Carriers for Targeted Drug Delivery System. Curr Top Med Chem 2021; 20:2472-2492. [PMID: 32962615 DOI: 10.2174/1568026620666200922113054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/25/2020] [Accepted: 04/25/2020] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane vesicles (MVs) playing important roles in various cellular and molecular functions in cell-to-cell signaling and transmitting molecular signals to adjacent as well as distant cells. The preserved cell membrane characteristics in MVs derived from live cells, give them great potential in biological applications. EVs are nanoscale particulates secreted from living cells and play crucial roles in several important cellular functions both in physiological and pathological states. EVs are the main elements in intercellular communication in which they serve as carriers for various endogenous cargo molecules, such as RNAs, proteins, carbohydrates, and lipids. High tissue tropism capacity that can be conveniently mediated by surface molecules, such as integrins and glycans, is a unique feature of EVs that makes them interesting candidates for targeted drug delivery systems. The cell-derived giant MVs have been exploited as vehicles for delivery of various anticancer agents and imaging probes and for implementing combinational phototherapy for targeted cancer treatment. Giant MVs can efficiently encapsulate therapeutic drugs and deliver them to target cells through the membrane fusion process to synergize photodynamic/photothermal treatment under light exposure. EVs can load diagnostic or therapeutic agents using different encapsulation or conjugation methods. Moreover, to prolong the blood circulation and enhance the targeting of the loaded agents, a variety of modification strategies can be exploited. This paper reviews the EVs-based drug delivery strategies in cancer therapy. Biological, pharmacokinetics and physicochemical characteristics, isolation techniques, engineering, and drug loading strategies of EVs are discussed. The recent preclinical and clinical progresses in applications of EVs and oncolytic virus therapy based on EVs, the clinical challenges and perspectives are discussed.
Collapse
Affiliation(s)
- Le-Yi Zhang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People's Hospital Chun’an
Branch), Hangzhou 311700, China
| | - Xue Yang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Hong Chen
- Department of Stomatology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou
Medical College, Hangzhou 310014, China
| | - Hong-Ying Pan
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China,Department of Infectious Diseases, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Zhi-Ming Hu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China,Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
13
|
Busatto S, Iannotta D, Walker SA, Di Marzio L, Wolfram J. A Simple and Quick Method for Loading Proteins in Extracellular Vesicles. Pharmaceuticals (Basel) 2021; 14:356. [PMID: 33924377 PMCID: PMC8069621 DOI: 10.3390/ph14040356] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular transport of biomolecular cargo in the body, making them promising delivery vehicles for bioactive compounds. Genetic engineering of producer cells has enabled encapsulation of therapeutic proteins in EVs. However, genetic engineering approaches can be expensive, time-consuming, and incompatible with certain EV sources, such as human plasma and bovine milk. The goal of this study was to develop a quick, versatile, and simple method for loading proteins in EVs post-isolation. Proteins, including CRISPR associated protein 9 (Cas9), were bound to cationic lipids that were further complexed with MDA-MB-231 cell-derived EVs through passive incubation. Size-exclusion chromatography was used to remove components that were not complexed with EVs. The ability of EVs to mediate intracellular delivery of proteins was compared to conventional methods, such as electroporation and commercial protein transfection reagents. The results indicate that EVs retain native features following protein-loading and obtain similar levels of intracellular protein delivery as conventional methods, but display less toxicity. This method opens up opportunities for rapid exploration of EVs for protein delivery.
Collapse
Affiliation(s)
- Sara Busatto
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Dalila Iannotta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Sierra A. Walker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
14
|
Ding L, Tang S, Wyatt TA, Knoell DL, Oupický D. Pulmonary siRNA delivery for lung disease: Review of recent progress and challenges. J Control Release 2021; 330:977-991. [PMID: 33181203 DOI: 10.1016/j.jconrel.2020.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/12/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Lung diseases are a leading cause of mortality worldwide and there exists urgent need for new therapies. Approval of the first siRNA treatments in humans has opened the door for further exploration of this therapeutic strategy for other disease states. Pulmonary delivery of siRNA-based biopharmaceuticals offers the potential to address multiple unmet medical needs in lung-related diseases because of the specific physiology of the lung and characteristic properties of siRNA. Inhalation-based siRNA delivery designed for efficient, targeted delivery to specific cells within the lung holds great promise. Efficient delivery of siRNA directly to the lung, however, is relatively complex. This review focuses on the barriers that impact pulmonary siRNA delivery and successful recent approaches to advance this field forward. We focus on the pulmonary barriers that affect siRNA delivery, the disease-dependent pathological changes and their role in pulmonary disease and impact on siRNA delivery, as well as the recent development on the pulmonary siRNA delivery systems.
Collapse
Affiliation(s)
- Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Todd A Wyatt
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Department of Veterans Affairs Nebraska, Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Daren L Knoell
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
15
|
Cevenini A, Celia C, Orrù S, Sarnataro D, Raia M, Mollo V, Locatelli M, Imperlini E, Peluso N, Peltrini R, De Rosa E, Parodi A, Del Vecchio L, Di Marzio L, Fresta M, Netti PA, Shen H, Liu X, Tasciotti E, Salvatore F. Liposome-Embedding Silicon Microparticle for Oxaliplatin Delivery in Tumor Chemotherapy. Pharmaceutics 2020; 12:pharmaceutics12060559. [PMID: 32560359 PMCID: PMC7355455 DOI: 10.3390/pharmaceutics12060559] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
Mesoporous silicon microparticles (MSMPs) can incorporate drug-carrying nanoparticles (NPs) into their pores. An NP-loaded MSMP is a multistage vector (MSV) that forms a Matryoshka-like structure that protects the therapeutic cargo from degradation and prevents its dilution in the circulation during delivery to tumor cells. We developed an MSV constituted by 1 µm discoidal MSMPs embedded with PEGylated liposomes containing oxaliplatin (oxa) which is a therapeutic agent for colorectal cancer (CRC). To obtain extra-small liposomes able to fit the 60 nm pores of MSMP, we tested several liposomal formulations, and identified two optimal compositions, with a prevalence of the rigid lipid 1,2-distearoyl-sn-glycero-3-phosphocholine and of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]. To improve the MSV assembly, we optimized the liposome-loading inside the MSMP and achieved a five-fold increase of the payload using an innovative lyophilization approach. This procedure also increased the load and limited dimensional changes of the liposomes released from the MSV in vitro. Lastly, we found that the cytotoxic efficacy of oxa-loaded liposomes and-oxa-liposome-MSV in CRC cell culture was similar to that of free oxa. This study increases knowledge about extra-small liposomes and their loading into porous materials and provides useful hints about alternative strategies for designing drug-encapsulating NPs.
Collapse
Affiliation(s)
- Armando Cevenini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Christian Celia
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Stefania Orrù
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Dipartimento di Scienze Motorie e del Benessere, Università “Parthenope”, 80133 Napoli, Italy
- IRCCS SDN, 80143 Napoli, Italy; (E.I.); (A.P.)
| | - Daniela Sarnataro
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
| | - Maddalena Raia
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Valentina Mollo
- Italian Institute of Technology@CRIB Center for Advanced Biomaterials for Health Care, 80125 Napoli, Italy; (V.M.); (P.A.N.)
| | - Marcello Locatelli
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
| | | | - Nicoletta Peluso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Rosa Peltrini
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Department of Respiratory Sciences, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Enrica De Rosa
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Alessandro Parodi
- IRCCS SDN, 80143 Napoli, Italy; (E.I.); (A.P.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Luigi Del Vecchio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annuzio”, 66100 Chieti, Italy; (C.C.); (M.L.); (L.D.M.)
| | - Massimo Fresta
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Campus Universitario “S. Venuta”, I-88100 Catanzaro, Italy;
| | - Paolo Antonio Netti
- Italian Institute of Technology@CRIB Center for Advanced Biomaterials for Health Care, 80125 Napoli, Italy; (V.M.); (P.A.N.)
- Department of Chemical, Materials & Industrial Production Engineering, University of Naples Federico II, 80125 Naples, Italy
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (E.D.R.); (H.S.); (X.L.)
| | - Ennio Tasciotti
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX 77030, USA
- Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Correspondence: (E.T.); (F.S.)
| | - Francesco Salvatore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (A.C.); (D.S.); (N.P.); (R.P.); (L.D.V.)
- CEINGE-Biotecnologie Avanzate S.c.a r.l., 80145 Napoli, Italy; (S.O.); (M.R.)
- Correspondence: (E.T.); (F.S.)
| |
Collapse
|
16
|
Zhu C, Zhu Y, Pan H, Chen Z, Zhu Q. Current Progresses of Functional Nanomaterials for Imaging Diagnosis and Treatment of Melanoma. Curr Top Med Chem 2019; 19:2494-2506. [PMID: 31642783 DOI: 10.2174/1568026619666191023130524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
Melanoma is a malignant skin tumor that results in poor disease prognosis due to unsuccessful
treatment options. During the early stages of tumor progression, surgery is the primary approach
that assures a good outcome. However, in the presence of metastasis, melanoma hasbecome almost
immedicable, since the tumors can not be removed and the disease recurs easily in a short period of
time. However, in recent years, the combination of nanomedicine and chemotherapeutic drugs has offered
promising solutions to the treatment of late-stage melanoma. Extensive studies have demonstrated
that nanomaterials and their advanced applications can improve the efficacy of traditional chemotherapeutic
drugs in order to overcome the disadvantages, such as drug resistance, low drug delivery rate and
reduced targeting to the tumor tissue. In the present review, we summarized the latest progress in imaging
diagnosis and treatment of melanoma using functional nanomaterials, including polymers,
liposomes, metal nanoparticles, magnetic nanoparticles and carbon-based nanoparticles. These
nanoparticles are reported widely in melanoma chemotherapy, gene therapy, immunotherapy, photodynamic
therapy, and hyperthermia.
Collapse
Affiliation(s)
- Congcong Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Yunjie Zhu
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Huijun Pan
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Zhongjian Chen
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Quangang Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
17
|
Walker S, Busatto S, Pham A, Tian M, Suh A, Carson K, Quintero A, Lafrence M, Malik H, Santana MX, Wolfram J. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics 2019; 9:8001-8017. [PMID: 31754377 PMCID: PMC6857056 DOI: 10.7150/thno.37097] [Citation(s) in RCA: 303] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring cell-secreted nanoparticles that play important roles in many physiological and pathological processes. EVs enable intercellular communication by serving as delivery vehicles for a wide range of endogenous cargo molecules, such as RNAs, proteins, carbohydrates, and lipids. EVs have also been found to display tissue tropism mediated by surface molecules, such as integrins and glycans, making them promising for drug delivery applications. Various methods can be used to load therapeutic agents into EVs, and additional modification strategies have been employed to prolong circulation and improve targeting. This review gives an overview of EV-based drug delivery strategies in cancer therapy.
Collapse
Affiliation(s)
- Sierra Walker
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sara Busatto
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Anthony Pham
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ming Tian
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Annie Suh
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Kelsey Carson
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Astrid Quintero
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Maria Lafrence
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Hanna Malik
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Moises X. Santana
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Joy Wolfram
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| |
Collapse
|
18
|
Klicks J, Maßlo C, Kluth A, Rudolf R, Hafner M. A novel spheroid-based co-culture model mimics loss of keratinocyte differentiation, melanoma cell invasion, and drug-induced selection of ABCB5-expressing cells. BMC Cancer 2019; 19:402. [PMID: 31035967 PMCID: PMC6489189 DOI: 10.1186/s12885-019-5606-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/12/2019] [Indexed: 01/18/2023] Open
Abstract
Background Different 3D-cell culture approaches with varying degrees of complexity have been developed to serve as melanoma models for drug testing or mechanistic studies. While these 3D-culture initiatives are already often superior to classical 2D approaches, they are either composed of only melanoma cells or they are so complex that the behavior of individual cell types is hard to understand, and often they are difficult to establish and expensive. Methods This study used low-attachment based generation of spheroids composed of up to three cell types. Characterization of cells and spheroids involved cryosectioning, immunofluorescence, FACS, and quantitative analyses. Statistical evaluation used one-way ANOVA with post-hoc Tukey test or Student’s t-test. Results The tri-culture model allowed to track cellular behavior in a cell-type specific manner and recapitulated different characteristics of early melanoma stages. Cells arranged into a collagen-IV rich fibroblast core, a ring of keratinocytes, and groups of highly proliferating melanoma cells on the outside. Regularly, some melanoma cells were also found to invade the fibroblast core. In the absence of melanoma cells, the keratinocyte ring stratified into central basal-like and peripheral, more differentiated cells. Conversely, keratinocyte differentiation was clearly reduced upon addition of melanoma cells. Treatment with the cytostatic drug, docetaxel, restored keratinocyte differentiation and induced apoptosis of external melanoma cells. Remaining intact external melanoma cells showed a significantly increased amount of ABCB5-immunoreactivity. Conclusions In the present work, a novel, simple spheroid-based melanoma tri-culture model composed of fibroblasts, keratinocytes, and melanoma cells was described. This model mimicked features observed in early melanoma stages, including loss of keratinocyte differentiation, melanoma cell invasion, and drug-induced increase of ABCB5 expression in external melanoma cells. Electronic supplementary material The online version of this article (10.1186/s12885-019-5606-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Klicks
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany.,Institute of Medical Technology, Mannheim University of Applied Sciences and Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christoph Maßlo
- RHEACELL GmbH & Co. KG, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Andreas Kluth
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany. .,Institute of Medical Technology, Mannheim University of Applied Sciences and Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Paul-Wittsack-Straße 10, 68163, Mannheim, Germany.,Institute of Medical Technology, Mannheim University of Applied Sciences and Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
19
|
Abstract
Most clinically approved drugs (primarily small molecules or antibodies) are rapidly cleared from circulation and distribute throughout the body. As a consequence, only a small portion of the dose accumulates at the target site, leading to low efficacy and adverse side effects. Therefore, new delivery strategies are necessary to increase organ and tissue-specific delivery of therapeutic agents. Nanoparticles provide a promising approach for prolonging the circulation time and improving the biodistribution of drugs. However, nanoparticles display several limitations, such as clearance by the immune systems and impaired diffusion in the tissue microenvironment. To overcome common nanoparticle limitations various functionalization and targeting strategies have been proposed. This review will discuss synthetic nanoparticle and extracellular vesicle delivery strategies that exploit organ-specific features to enhance drug accumulation at the target site.
Collapse
|
20
|
Abstract
Nanotechnology offers new solutions for the development of cancer therapeutics that display improved efficacy and safety. Although several nanotherapeutics have received clinical approval, the most promising nanotechnology applications for patients still lie ahead. Nanoparticles display unique transport, biological, optical, magnetic, electronic, and thermal properties that are not apparent on the molecular or macroscale, and can be utilized for therapeutic purposes. These characteristics arise because nanoparticles are in the same size range as the wavelength of light and display large surface area to volume ratios. The large size of nanoparticles compared to conventional chemotherapeutic agents or biological macromolecule drugs also enables incorporation of several supportive components in addition to active pharmaceutical ingredients. These components can facilitate solubilization, protection from degradation, sustained release, immunoevasion, tissue penetration, imaging, targeting, and triggered activation. Nanoparticles are also processed differently in the body compared to conventional drugs. Specifically, nanoparticles display unique hemodynamic properties and biodistribution profiles. Notably, the interactions that occur at the bio-nano interface can be exploited for improved drug delivery. This review discusses successful clinically approved cancer nanodrugs as well as promising candidates in the pipeline. These nanotherapeutics are categorized according to whether they predominantly exploit multifunctionality, unique electromagnetic properties, or distinct transport characteristics in the body. Moreover, future directions in nanomedicine such as companion diagnostics, strategies for modifying the microenvironment, spatiotemporal nanoparticle transitions, and the use of extracellular vesicles for drug delivery are also explored.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida 32224, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
21
|
Mishra H, Mishra PK, Ekielski A, Jaggi M, Iqbal Z, Talegaonkar S. Melanoma treatment: from conventional to nanotechnology. J Cancer Res Clin Oncol 2018; 144:2283-2302. [PMID: 30094536 DOI: 10.1007/s00432-018-2726-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 07/30/2018] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Melanoma is the most serious form of skin cancer causing most of the skin cancer-related deaths. The incidence of melanoma has risen so dramatically over past few years that no other solid or blood malignancy comes close to it in terms of increased incidence. The main problem associated with the treatment of melanoma is low response rate to the existing treatment modalities, which in turn is due to the incomplete response by chemotherapeutic agents and inherent resistance of melanoma cells. MATERIALS AND METHODS Conventional therapeutic strategies, as well as, recent literature on melanoma have been thoroughly studied. This review summarizes the base of anti-melanoma treatment with conventional chemotherapeutic drugs, followed by an account of recent studies which explored the potential of nanotechnology and newer strategies and agents in melanoma treatment. CONCLUSION Although melanoma is curable if detected in its early localized form, metastatic melanoma continues to be a therapeutic challenge. Metastatic melanoma has a very poor prognosis and conventional therapies have not improved the outcomes of the treatment so far. For this reason, newer combinations of anti-melanoma drugs and newer strategies utilizing nanotechnology have been constantly explored.
Collapse
Affiliation(s)
- Harshita Mishra
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Pawan K Mishra
- Department of Wood Processing Technologies, Mendel University in Brno, Brno, Czech Republic
| | - Adam Ekielski
- Department of Production Management and Engineering, Faculty of Production Engineering, Warsaw University of Life Sciences, Warsaw, Poland
| | - Manu Jaggi
- Dabur Research Foundation, Ghaziabad, India
| | - Zeenat Iqbal
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sushama Talegaonkar
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Government of NCT of Delhi, New Delhi, India.
| |
Collapse
|
22
|
Luo Z, Jiang L, Ding C, Hu B, Loh XJ, Li Z, Wu Y. Surfactant Free Delivery of Docetaxel by Poly[(R)-3-hydroxybutyrate-(R)-3-hydroxyhexanoate]-Based Polymeric Micelles for Effective Melanoma Treatments. Adv Healthc Mater 2018; 7:e1801221. [PMID: 30398017 DOI: 10.1002/adhm.201801221] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/23/2018] [Indexed: 12/13/2022]
Abstract
Docetaxel (DTX) is a new semisynthetic chemical in the taxoid family and serves a wide spectrum of chemotherapeutics. Current commercial formulation of DTX is based on the addition of the nonionic surfactants (i.e., ethanol and Tween 80), which are reported to cause severe hemolysis, hypersensitivity reactions, or neurotoxic toxicity and greatly hinders patient tolerance or compliance. In this report, a novel low-toxic, biodegradable, and amphiphilic poly[(R)-3-hydroxybutyrate-(R)-3-hydroxyhexanoate] (PHBHx)-based polyurethane (a copolymer made of hydrophobic PHBHx with biocompatible D-3-hydroxybutyric acid as degradation product, thermosensitive polypropylene glycol (PPG), and hydrophilic polyethylene glycol (PEG) segments) with nanosized micelle formation ability to encapsulate DTX, as a surfactant free formulation, is reported. Interestingly, this DTX-loaded poly(PHBHx/PEG/PPG urethane) micelle formulation with >90% drug loading efficiency shows significantly improved DTX solubility in aqueous medium, reduced hemolysis for better blood compatibility, and increased drug uptake in A375 melanoma cells, which provides the possibility of systematic delivery of DTX. As a proof-of-concept, an A375 melanoma xenograft mouse model is established to verify the therapeutic effect of this DTX-loaded poly(PHBHx/PEG/PPG urethane) micelle formulation, indicating the promising application of PHBHx-based polymeric nanosized micelle as a surfactant free formulation of chemotherapeutics which might greatly be beneficial for controllable delivery of pharmaceutics and cancer therapy.
Collapse
Affiliation(s)
- Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress BiologySchool of Pharmaceutical SciencesXiamen University Xiamen 361102 P. R. China
| | - Lu Jiang
- Institute of Materials Research and EngineeringA*STAR (Agency for Science, Technology and Research) 2 Fusionopolis Way, Innovis, #08‐03 Singapore 138634 Singapore
| | - Chizhu Ding
- College of ScienceHuazhong Agricultural University Wuhan 430074 P. R. China
| | - Benhui Hu
- School of Biomedical Engineering and InformaticsNanjing Medical University Nanjing 211166 P. R. China
| | - Xian Jun Loh
- Institute of Materials Research and EngineeringA*STAR (Agency for Science, Technology and Research) 2 Fusionopolis Way, Innovis, #08‐03 Singapore 138634 Singapore
| | - Zibiao Li
- Institute of Materials Research and EngineeringA*STAR (Agency for Science, Technology and Research) 2 Fusionopolis Way, Innovis, #08‐03 Singapore 138634 Singapore
| | - Yun‐Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress BiologySchool of Pharmaceutical SciencesXiamen University Xiamen 361102 P. R. China
| |
Collapse
|
23
|
Mattu C, Brachi G, Menichetti L, Flori A, Armanetti P, Ranzato E, Martinotti S, Nizzero S, Ferrari M, Ciardelli G. Alternating block copolymer-based nanoparticles as tools to modulate the loading of multiple chemotherapeutics and imaging probes. Acta Biomater 2018; 80:341-351. [PMID: 30236799 DOI: 10.1016/j.actbio.2018.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/30/2018] [Accepted: 09/15/2018] [Indexed: 12/30/2022]
Abstract
Cancer therapy often relies on the combined action of different molecules to overcome drug resistance and enhance patient outcome. Combined strategies relying on molecules with different pharmacokinetics often fail due to the lack of concomitant tumor accumulation and, thus, to the loss of synergistic effect. Due to their ability to enhance treatment efficiency, improve drug pharmacokinetics, and reduce adverse effects, polymer nanoparticles (PNPs) have been widely investigated as co-delivery vehicles for cancer therapies. However, co-encapsulation of different drugs and probes in PNPs requires a flexible polymer platform and a tailored particle design, in which both the bulk and surface properties of the carriers are carefully controlled. In this work, we propose a core-shell PNP design based on a polyurethane (PUR) core and a phospholipid external surface. The modulation of the hydrophilic/hydrophobic balance of the PUR core enhanced the encapsulation of two chemotherapeutics with dramatically different water solubility (Doxorubicin hydrochloride, DOXO and Docetaxel, DCTXL) and of Iron Oxide Nanoparticles for MRI imaging. The outer shell remained unchanged among the platforms, resulting in un-modified cellular uptake and in vivo biodistribution. We demonstrate that the choice of PUR core allowed a high entrapment efficiency of all drugs, superior or comparable to previously reported results, and that higher core hydrophilicity enhances the loading efficiency of the hydrophilic DOXO and the MRI contrast effect. Moreover, we show that changing the PUR core did not alter the surface properties of the carriers, since all particles showed a similar behavior in terms of cell internalization and in vivo biodistribution. We also show that PUR PNPs have high passive tumor accumulation and that they can efficient co-deliver the two drugs to the tumor, reaching an 11-fold higher DOXO/DCTXL ratio in tumor as compared to free drugs. STATEMENT OF SIGNIFICANCE: Exploiting the synergistic action of multiple chemotherapeutics is a promising strategy to improve the outcome of cancer patients, as different agents can simultaneously engage different features of tumor cells and/or their microenvironment. Unfortunately, the choice is limited to drugs with similar pharmacokinetics that can concomitantly accumulate in tumors. To expand the spectrum of agents that can be delivered in combination, we propose a multi-compartmental core-shell nanoparticles approach, in which the core is made of biomaterials with high affinity for drugs of different physical properties. We successfully co-encapsulated Doxorubicin Hydrochloride, Docetaxel, and contrast agents and achieved a significantly higher concomitant accumulation in tumor versus free drugs, demonstrating that nanoparticles can improve synergistic cancer chemotherapy.
Collapse
Affiliation(s)
- C Mattu
- Politecnico di Torino, DIMEAS C.so Duca degli Abruzzi 24, 10129 Torino, Italy; Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - G Brachi
- Politecnico di Torino, DIMEAS C.so Duca degli Abruzzi 24, 10129 Torino, Italy; Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA
| | - L Menichetti
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi, 1 56124 Pisa, Italy; Fondazione Regione Toscana G. Monasterio, Via Giuseppe Moruzzi 1, Pisa 56124, Italy
| | - A Flori
- Fondazione Regione Toscana G. Monasterio, Via Giuseppe Moruzzi 1, Pisa 56124, Italy
| | - P Armanetti
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi, 1 56124 Pisa, Italy
| | - E Ranzato
- DiSIT-Dipartimento di Scienze e Innovazione Tecnologica, University of Piemonte Orientale, piazza Sant'Eusebio 5, Vercelli 13100, Italy
| | - S Martinotti
- DiSIT-Dipartimento di Scienze e Innovazione Tecnologica, University of Piemonte Orientale, Viale Teresa Michel 11, Alessandria 15121, Italy
| | - S Nizzero
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA; Applied Physics Graduate Program, Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
| | - M Ferrari
- Department of Nanomedicine, Houston Methodist Hospital Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - G Ciardelli
- Politecnico di Torino, DIMEAS C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
24
|
Tieu T, Alba M, Elnathan R, Cifuentes‐Rius A, Voelcker NH. Advances in Porous Silicon–Based Nanomaterials for Diagnostic and Therapeutic Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800095] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- Prof. N. H. Voelcker Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| |
Collapse
|
25
|
Wintzheimer S, Granath T, Oppmann M, Kister T, Thai T, Kraus T, Vogel N, Mandel K. Supraparticles: Functionality from Uniform Structural Motifs. ACS NANO 2018; 12:5093-5120. [PMID: 29763295 DOI: 10.1021/acsnano.8b00873] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Under the right process conditions, nanoparticles can cluster together to form defined, dispersed structures, which can be termed supraparticles. Controlling the size, shape, and morphology of such entities is a central step in various fields of science and technology, ranging from colloid chemistry and soft matter physics to powder technology and pharmaceutical and food sciences. These diverse scientific communities have been investigating formation processes and structure/property relations of such supraparticles under completely different boundary conditions. On the fundamental side, the field is driven by the desire to gain maximum control of the assembly structures using very defined and tailored colloidal building blocks, whereas more applied disciplines focus on optimizing the functional properties from rather ill-defined starting materials. With this review article, we aim to provide a connecting perspective by outlining fundamental principles that govern the formation and functionality of supraparticles. We discuss the formation of supraparticles as a result of colloidal properties interplaying with external process parameters. We then outline how the structure of the supraparticles gives rise to diverse functional properties. They can be a result of the structure itself (emergent properties), of the colocalization of different, functional building blocks, or of coupling between individual particles in close proximity. Taken together, we aim to establish structure-property and process-structure relationships that provide unifying guidelines for the rational design of functional supraparticles with optimized properties. Finally, we aspire to connect the different disciplines by providing a categorized overview of the existing, diverging nomenclature of seemingly similar supraparticle structures.
Collapse
Affiliation(s)
- Susanne Wintzheimer
- Fraunhofer Institute for Silicate Research, ISC , Neunerplatz 2 , 97082 Würzburg , Germany
| | - Tim Granath
- Chair of Chemical Technology of Materials Synthesis , University Würzburg , Röntgenring 11 , 97070 Würzburg , Germany
| | - Maximilian Oppmann
- Fraunhofer Institute for Silicate Research, ISC , Neunerplatz 2 , 97082 Würzburg , Germany
| | - Thomas Kister
- INM-Leibniz Institute for New Materials , Campus D2 2, 66123 Saarbrücken , Germany
| | - Thibaut Thai
- INM-Leibniz Institute for New Materials , Campus D2 2, 66123 Saarbrücken , Germany
| | - Tobias Kraus
- INM-Leibniz Institute for New Materials , Campus D2 2, 66123 Saarbrücken , Germany
- Colloid and Interface Chemistry , Saarland University , Campus D2 2, 66123 Saarbrücken , Germany
| | - Nicolas Vogel
- Institute of Particle Technology , Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) , Haberstrasse 9A , 91058 Erlangen , Germany
| | - Karl Mandel
- Fraunhofer Institute for Silicate Research, ISC , Neunerplatz 2 , 97082 Würzburg , Germany
- Chair of Chemical Technology of Materials Synthesis , University Würzburg , Röntgenring 11 , 97070 Würzburg , Germany
| |
Collapse
|
26
|
Pelt J, Busatto S, Ferrari M, Thompson EA, Mody K, Wolfram J. Chloroquine and nanoparticle drug delivery: A promising combination. Pharmacol Ther 2018; 191:43-49. [PMID: 29932886 DOI: 10.1016/j.pharmthera.2018.06.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Clinically approved cancer therapies include small molecules, antibodies, and nanoparticles. There has been major progress in the treatment of several cancer types over recent decades. However, many challenges remain for optimal use of conventional and nanoparticle-based therapies in oncology including poor drug delivery, rapid clearance, and drug resistance. The antimalarial agent chloroquine has been found to mitigate some of these challenges by modulating cancer cells and the tissue microenvironment. Particularly, chloroquine was recently found to reduce immunological clearance of nanoparticles by resident macrophages in the liver, leading to increased tumor accumulation of nanodrugs. Additionally, chloroquine has been shown to improve drug delivery and efficacy through normalization of tumor vasculature and suppression of several oncogenic and stress-tolerance pathways, such as autophagy, that protect cancer cells from cytotoxic agents. This review will discuss the use of chloroquine as combination therapy to improve cancer treatment.
Collapse
Affiliation(s)
- Joe Pelt
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Florida State University, Tallahassee, FL 32306, USA
| | - Sara Busatto
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Molecular and Translational Medicine, University of Brescia, Brescia 25133, Italy.
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kabir Mody
- Division of Hematology/Oncology, Mayo Clinic Cancer Center, Mayo Clinic Florida, Jacksonville, FL 32224, USA.
| | - Joy Wolfram
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
27
|
Li W, Liu Z, Fontana F, Ding Y, Liu D, Hirvonen JT, Santos HA. Tailoring Porous Silicon for Biomedical Applications: From Drug Delivery to Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1703740. [PMID: 29534311 DOI: 10.1002/adma.201703740] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/16/2017] [Indexed: 05/24/2023]
Abstract
In the past two decades, porous silicon (PSi) has attracted increasing attention for its potential biomedical applications. With its controllable geometry, tunable nanoporous structure, large pore volume/high specific surface area, and versatile surface chemistry, PSi shows significant advantages over conventional drug carriers. Here, an overview of recent progress in the use of PSi in drug delivery and cancer immunotherapy is presented. First, an overview of the fabrication of PSi with various geometric structures is provided, with particular focus on how the unique geometry of PSi facilitates its biomedical applications, especially for drug delivery. Second, surface chemistry and modification of PSi are discussed in relation to the strengthening of its performance in drug delivery and bioimaging. Emerging technologies for engineering PSi-based composites are then summarized. Emerging PSi advances in the context of cancer immunotherapy are also highlighted. Overall, very promising research results encourage further exploration of PSi for biomedical applications, particularly in drug delivery and cancer immunotherapy, and future translation of PSi into clinical applications.
Collapse
Affiliation(s)
- Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yaping Ding
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Dongfei Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| | - Jouni T Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
28
|
Su Y, Wang K, Li Y, Song W, Xin Y, Zhao W, Tian J, Ren L, Lu L. Sorafenib-loaded polymeric micelles as passive targeting therapeutic agents for hepatocellular carcinoma therapy. Nanomedicine (Lond) 2018; 13:1009-1023. [PMID: 29630448 DOI: 10.2217/nnm-2018-0046] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM The clinical application of sorafenib is limited because of its hydrophobicity, low bioavailability and unsatisfying treatment effect. Therefore, sorafenib-loaded PEG-poly (ε-caprolactone) micelles (SF micelles) were fabricated for sorafenib delivery. MATERIALS & METHODS In vitro assays investigated the solubility, dispersity, stability, cytotoxicity and uptake capacity of SF micelles. In vivo biodistribution and therapeutic effects were studied using HepG2-Luc tumor-bearing mice. RESULTS SF micelles had a regular spherical structure with good water solubility. In vivo imaging results showed PEG-poly (ε-caprolactone) micelles could elevate the sorafenib concentration in tumor tissues. Meanwhile, SF micelles exhibited higher tumor growth inhibition in vivo. CONCLUSION SF micelles might be a potential drug delivery system, which could enhance the therapeutic effects of sorafenib.
Collapse
Affiliation(s)
- Yanhong Su
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Center of Intervention Radiology, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai 519000, China.,CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management & Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management & Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong Li
- Center of Intervention Radiology, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai 519000, China
| | - Wenjing Song
- School of Materials Science & Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yongjie Xin
- Center of Intervention Radiology, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai 519000, China
| | - Wei Zhao
- Center of Intervention Radiology, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai 519000, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management & Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Ren
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,School of Materials Science & Engineering, South China University of Technology, Guangzhou 510641, China
| | - Ligong Lu
- School of Medicine, South China University of Technology, Guangzhou 510006, China.,Center of Intervention Radiology, Zhuhai Precision Medicine Center, Zhuhai People's Hospital, Zhuhai 519000, China
| |
Collapse
|
29
|
Shen J, Zhang W, Qi R, Mao ZW, Shen H. Engineering functional inorganic-organic hybrid systems: advances in siRNA therapeutics. Chem Soc Rev 2018; 47:1969-1995. [PMID: 29417968 PMCID: PMC5861001 DOI: 10.1039/c7cs00479f] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer treatment still faces a lot of obstacles such as tumor heterogeneity, drug resistance and systemic toxicities. Beyond the traditional treatment modalities, exploitation of RNA interference (RNAi) as an emerging approach has immense potential for the treatment of various gene-caused diseases including cancer. The last decade has witnessed enormous research and achievements focused on RNAi biotechnology. However, delivery of small interference RNA (siRNA) remains a key challenge in the development of clinical RNAi therapeutics. Indeed, functional nanomaterials play an important role in siRNA delivery, which could overcome a wide range of sequential physiological and biological obstacles. Nanomaterial-formulated siRNA systems have potential applications in protection of siRNA from degradation, improving the accumulation in the target tissues, enhancing the siRNA therapy and reducing the side effects. In this review, we explore and summarize the role of functional inorganic-organic hybrid systems involved in the siRNA therapeutic advancements. Additionally, we gather the surface engineering strategies of hybrid systems to optimize for siRNA delivery. Major progress in the field of inorganic-organic hybrid platforms including metallic/non-metallic cores modified with organic shells or further fabrication as the vectors for siRNA delivery is discussed to give credit to the interdisciplinary cooperation between chemistry, pharmacy, biology and medicine.
Collapse
Affiliation(s)
- Jianliang Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China and Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Science, Wenzhou, 325001, China and Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Wei Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Ruogu Qi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY10065, USA
| |
Collapse
|
30
|
Borrelli DA, Yankson K, Shukla N, Vilanilam G, Ticer T, Wolfram J. Extracellular vesicle therapeutics for liver disease. J Control Release 2018; 273:86-98. [PMID: 29373816 DOI: 10.1016/j.jconrel.2018.01.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are endogenous nanoparticles that play important roles in intercellular communication. Unmodified and engineered EVs can be utilized for therapeutic purposes. For instance, mesenchymal stem cell (MSC)-derived EVs have shown promise for tissue repair, while drug-loaded EVs have the potential to be used for cancer treatment. The liver is an ideal target for EV therapy due to the intrinsic regenerative capacity of hepatic tissue and the tropism of systemically injected nanovesicles for this organ. This review will give an overview of the potential of EV therapeutics in liver disease. Specifically, the mechanisms by which MSC-EVs induce liver repair will be covered. Moreover, the use of drug-loaded EVs for the treatment of hepatocellular carcinoma will also be discussed. Although there are several challenges associated with the clinical translation of EVs, these biological nanoparticles represent a promising new therapeutic modality for liver disease.
Collapse
Affiliation(s)
- David A Borrelli
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kiera Yankson
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Neha Shukla
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - George Vilanilam
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Taylor Ticer
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Joy Wolfram
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; Wenzhou Institute of Biomaterials and Engineering, Ningbo Institute of Industrial Technology, Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
31
|
Nanomedicine, an emerging therapeutic strategy for oral cancer therapy. Oral Oncol 2017; 76:1-7. [PMID: 29290280 DOI: 10.1016/j.oraloncology.2017.11.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/06/2017] [Accepted: 11/12/2017] [Indexed: 01/12/2023]
Abstract
Oral cavity and oropharyngeal carcinomas (oral cancer) represents a significant cause of morbidity and mortality. Despite efforts in improving early diagnosis and treatment, the 5-year survival rate of advanced stage of the disease is less than 63%. The field of nanomedicine has offered promising diagnostic and therapeutic advances in cancer. Indeed, several platforms have been clinically approved for cancer therapy, while other promising systems are undergoing exploration in clinical trials. With its ability to deliver drugs, nucleic acids, and MRI contrast agents with high efficiency, nanomedicine platforms offer the potential to improve drug efficacy and tolerability. The aim of the present mini-review is to summarize the current preclinical status of nanotechnology systems for oral cancer therapy. The nanoplatforms for delivery of chemopreventive agents presented herein resulted in significantly higher anti-tumor activity than free forms of the drug, even against a chemo-resistant cell line. Impressive results have also been obtained using nanoparticles to deliver chemotherapeutics, resulting in reduced toxicity both in vitro and in vivo. Nanoparticles have also led to improvements in efficacy of photodynamic therapies through the development of targeted magnetic nanoparticles. Finally, gene therapy using nanoparticles demonstrated promising results specifically with regards to inhibition of gene expression. Of the few in vivo studies that have been reported, many of these used animal models with several limitations, which will be discussed herein. Lastly, we will discuss several future perspectives in oral cancer nanoparticle-based therapy and the development of appropriate animal models, distinguishing between oral cavity and oropharyngeal carcinoma.
Collapse
|
32
|
Youngren-Ortiz SR, Chougule MB. The Daniel K. Inouye College of Pharmacy Scripts: Targeted Nanocarrier Based Systems for the Treatment of Lung Cancer. HAWAI'I JOURNAL OF MEDICINE & PUBLIC HEALTH : A JOURNAL OF ASIA PACIFIC MEDICINE & PUBLIC HEALTH 2017; 76:318-325. [PMID: 29164017 PMCID: PMC5694976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In Hawai'i, lung cancer is among the top cancers diagnosed and a leading cause of death. Despite current understanding and modern surgery, radiology, and chemotherapy techniques, the survival of those suffering from lung cancer remains low. Current anticancer drugs have poor tumor tissue selectivity and toxicity issues that contribute to their overall low efficacy, detrimental effects to normal tissues, and drug resistance. A potential way of mitigating cancer is through RNA interference (RNAi) by the delivery of small interfering RNA (siRNA) to target select proteins or genes involved in cancer progression, known as oncoproteins or oncogenes, respectively. However, the clinical utility of delivering unformulated siRNA has been hindered due to poor cell penetration, nonspecific effects, rapid degradation, and short half-life. As an alternate for conventional chemotherapy, nanoparticles (AKA nanocarriers) may be designed to localize within the tumor environment and increase targeted cell internalization, thus reducing systemic adverse effects and increasing efficacy. Nanoparticles play important roles in drug delivery and have been widely studied for cancer therapy and diagnostics, termed collectively as theranostics. Nanoparticles composed of natural and artificial polymers, proteins, lipids, metals, and carbon-based materials have been developed for the delivery of siRNA. Cancer targeting has been improved by nanoparticle surface modification or conjugation with biomolecules that are attracted to or stimulate therapeutic agent release within cancer tissues or cells. In this mini-review article, we present recent progress in nanocarrier-mediated siRNA delivery systems that include lipid, polymer, metallic and carbon-based nanoparticles for lung cancer therapy.
Collapse
Affiliation(s)
- Susanne R Youngren-Ortiz
- Translational Drug Delivery Research Laboratory (DDR), Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI (SRY-O, MBC)
| | - Mahavir B Chougule
- Translational Drug Delivery Research Laboratory (DDR), Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI (SRY-O, MBC)
| |
Collapse
|
33
|
Wolfram J, Nizzero S, Liu H, Li F, Zhang G, Li Z, Shen H, Blanco E, Ferrari M. A chloroquine-induced macrophage-preconditioning strategy for improved nanodelivery. Sci Rep 2017; 7:13738. [PMID: 29062065 PMCID: PMC5653759 DOI: 10.1038/s41598-017-14221-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 10/06/2017] [Indexed: 11/09/2022] Open
Abstract
Site-specific localization is critical for improving the therapeutic efficacy and safety of drugs. Nanoparticles have emerged as promising tools for localized drug delivery. However, over 90% of systemically injected nanocarriers typically accumulate in the liver and spleen due to resident macrophages that form the mononuclear phagocyte system. In this study, the clinically approved antimalarial agent chloroquine was shown to reduce nanoparticle uptake in macrophages by suppressing endocytosis. Pretreatment of mice with a clinically relevant dose of chloroquine substantially decreased the accumulation of liposomes and silicon particles in the mononuclear phagocyte system and improved tumoritropic and organotropic delivery. The novel use of chloroquine as a macrophage-preconditioning agent presents a straightforward approach for addressing a major barrier in nanomedicine. Moreover, this priming strategy has broad applicability for improving the biodistribution and performance of particulate delivery systems. Ultimately, this study defines a paradigm for the combined use of macrophage-modulating agents with nanotherapeutics for improved site-specific delivery.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Transplantation, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Sara Nizzero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Applied Physics Graduate Program, Rice University, Houston, TX, 77005, USA
| | - Haoran Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Feng Li
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Guodong Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Zheng Li
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
34
|
Oshima G, Guo N, He C, Stack ME, Poon C, Uppal A, Wightman SC, Parekh A, Skowron KB, Posner MC, Lin W, Khodarev NN, Weichselbaum RR. In Vivo Delivery and Therapeutic Effects of a MicroRNA on Colorectal Liver Metastases. Mol Ther 2017; 25:1588-1595. [PMID: 28457664 PMCID: PMC5498809 DOI: 10.1016/j.ymthe.2017.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 11/16/2022] Open
Abstract
Multiple therapeutic agents are typically used in concert to effectively control metastatic tumors. Recently, we described microRNAs that are associated with the oligometastatic state, in which a limited number of metastatic tumors progress to more favorable outcomes. Here, we report the effective delivery of an oligometastatic microRNA (miR-655-3p) to colorectal liver metastases using nanoscale coordination polymers (NCPs). The NCPs demonstrated a targeted and prolonged distribution of microRNAs to metastatic liver tumors. Tumor-targeted microRNA miR-655-3p suppressed tumor growth when co-delivered with oxaliplatin, suggesting additive or synergistic interactions between microRNAs and platinum drugs. This is the first known example of systemically administered nanoparticles delivering an oligometastatic microRNA to advanced metastatic liver tumors and demonstrating tumor-suppressive effects. Our results suggest a potential therapeutic strategy for metastatic liver disease by the co-delivery of microRNAs and conventional cytotoxic agents using tumor-specific NCPs.
Collapse
Affiliation(s)
- Go Oshima
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Nining Guo
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA; Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Chunbai He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Melinda E Stack
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Christopher Poon
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Abhineet Uppal
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Sean C Wightman
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Akash Parekh
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Kinga B Skowron
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Mitchell C Posner
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA.
| | - Nikolai N Khodarev
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA.
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
35
|
Papa AL, Korin N, Kanapathipillai M, Mammoto A, Mammoto T, Jiang A, Mannix R, Uzun O, Johnson C, Bhatta D, Cuneo G, Ingber DE. Ultrasound-sensitive nanoparticle aggregates for targeted drug delivery. Biomaterials 2017; 139:187-194. [PMID: 28618348 DOI: 10.1016/j.biomaterials.2017.06.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 05/25/2017] [Accepted: 06/03/2017] [Indexed: 12/22/2022]
Abstract
Here we describe injectable, ultrasound (US)-responsive, nanoparticle aggregates (NPAs) that disintegrate into slow-release, nanoscale, drug delivery systems, which can be targeted to selective sites by applying low-energy US locally. We show that, unlike microbubble based drug carriers which may suffer from stability problems, the properties of mechanical activated NPAs, composed of polymer nanoparticles, can be tuned by properly adjusting the polymer molecular weight, the size of the nanoparticle precursors as well as the percentage of excipient utilized to hold the NPA together. We then apply this concept to practice by fabricating NPAs composed of nanoparticles loaded with Doxorubicin (Dox) and tested their ability to treat tumors via ultrasound activation. Mouse studies demonstrated significantly increased efficiency of tumor targeting of the US-activated NPAs compared to PLGA nanoparticle controls (with or without US applied) or intact NPAs. Importantly, when the Dox-loaded NPAs were injected and exposed to US energy locally, this increased ability to concentrate nanoparticles at the tumor site resulted in a significantly greater reduction in tumor volume compared to tumors treated with a 20-fold higher dose of the free drug.
Collapse
Affiliation(s)
- Anne-Laure Papa
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Netanel Korin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | | | - Akiko Mammoto
- Vascular Biology Program and Dept. of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Tadanori Mammoto
- Vascular Biology Program and Dept. of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amanda Jiang
- Vascular Biology Program and Dept. of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Vascular Biology Program and Dept. of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Oktay Uzun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Christopher Johnson
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Deen Bhatta
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Garry Cuneo
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Vascular Biology Program and Dept. of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
36
|
Dai W, Wang X, Song G, Liu T, He B, Zhang H, Wang X, Zhang Q. Combination antitumor therapy with targeted dual-nanomedicines. Adv Drug Deliv Rev 2017; 115:23-45. [PMID: 28285944 DOI: 10.1016/j.addr.2017.03.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
Combination therapy is one of the important treatment strategies for cancer at present. However, the outcome of current combination therapy based on the co-administration of conventional dosage forms is suboptimal, due to the short half-lives of chemodrugs, their deficient tumor selectivity and so forth. Nanotechnology-based targeted delivery systems show great promise in addressing the associated problems and providing superior therapeutic benefits. In this review, we focus on the combination of therapeutic strategies between different nanomedicines or drug-loaded nanocarriers, rather than the co-delivery of different drugs via a single nanocarrier. We introduce the general concept of various targeting strategies of nanomedicines, present the principles of combination antitumor therapy with dual-nanomedicines, analyze their advantages and limitations compared with co-delivery strategies, and overview the recent advances of combination therapy based on targeted nanomedicines. Finally, we reviewed the challenges and future perspectives regarding the selection of therapeutic agents, targeting efficiency and the gap between the preclinical and clinical outcome.
Collapse
Affiliation(s)
- Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoyou Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Ge Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Tongzhou Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing 100191, China.
| |
Collapse
|
37
|
Venuta A, Wolfram J, Shen H, Ferrari M. Post-nano strategies for drug delivery: Multistage porous silicon microvectors. J Mater Chem B 2016; 5:207-219. [PMID: 28670454 DOI: 10.1039/c6tb01978a] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanodelivery systems usually improve the biodistribution of drugs, leading to reduced side effects and enhanced therapeutic efficacy. However, only a small portion of the injected nanoparticle dose accumulates in pathological tissue. Challenges in drug delivery arise due to a multitude of transport obstacles in the body, including the endothelium, the extracellular matrix, and the cell membrane. In general, nanoparticles are designed to overcome only a few biological barriers, making them inadequate for localized drug delivery. Accordingly, a multifunctional and multicomponent systems are required to effectively address a wide variety of transport obstacles. A suitable approach to obtain high levels of multifunctionality is to bring together the nanoscale with the microscale, resulting in post-nano strategies for drug delivery. This review discusses several such post-nano approaches, with an emphasis on the multistage vector (MSV) platform. The MSV consists of three components on different spatial scales, each intended to address biological barriers that exist in a specific compartment in the body. The first stage vector is a microparticle that is designed to navigate in the vascular compartment. The second stage vector consists of nanoparticles that are released from the microparticle into the tissue interstitium, where they address biological barriers in extracellular and intracellular compartments. The final component of the system is a small molecule therapeutic agent. A new generation of microparticle-based strategies with expanded applications has recently been developed, including injectable nanoparticle generators and silicon particles for immunotherapy. Notably, the advantage of incorporating microstructures in drug delivery vehicles is apparent from the observation that superior functionality only appears on the microscale, highlighting the inherent functional limitations of nanostructures.
Collapse
Affiliation(s)
- Alessandro Venuta
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA.,Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
38
|
Turner CT, Hasanzadeh Kafshgari M, Melville E, Delalat B, Harding F, Mäkilä E, Salonen JJ, Cowin AJ, Voelcker NH. Delivery of Flightless I siRNA from Porous Silicon Nanoparticles Improves Wound Healing in Mice. ACS Biomater Sci Eng 2016; 2:2339-2346. [DOI: 10.1021/acsbiomaterials.6b00550] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Christopher T. Turner
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Morteza Hasanzadeh Kafshgari
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Elizabeth Melville
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Bahman Delalat
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Francis Harding
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Ermei Mäkilä
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jarno J. Salonen
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Allison J. Cowin
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Nicolas H. Voelcker
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
39
|
Mi Y, Wolfram J, Mu C, Liu X, Blanco E, Shen H, Ferrari M. Enzyme-responsive multistage vector for drug delivery to tumor tissue. Pharmacol Res 2016; 113:92-99. [PMID: 27546164 DOI: 10.1016/j.phrs.2016.08.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/02/2016] [Accepted: 08/18/2016] [Indexed: 10/21/2022]
Abstract
Various nanodelivery systems have been designed to release therapeutic agents upon contact with specific enzymes. However, enzyme-triggered release typically takes place in the tissue interstitium, thereby resulting in the extracellular delivery of drugs. Here, we have designed an enzyme-stimulated multistage vector (ESMSV), which enables stimulus-triggered release of drug-encapsulated nanoparticles from a microparticle. Specifically, polymeric nanoparticles with a surface matrix metalloproteinase-2 (MMP2) peptide substrate were conjugated to the surface of porous silicon microparticles. In the presence of MMP2, the polymeric nanoparticles were released into the tumor interstitium. This platform can be used to attain triggered drug release, while simultaneously facilitating the cellular internalization of drugs. The results indicate that nanoparticle release was MMP2-specific and resulted in improved intracellular uptake of hydrophobic agents in the presence of MMP2. Furthermore, in a mouse model of melanoma lung metastasis, systemic delivery of ESMSVs caused a substantial increase in intracellular accumulation of agents in cancer cells in comparison to delivery with non-stimulus-responsive particles.
Collapse
Affiliation(s)
- Yu Mi
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Chaofeng Mu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
40
|
Scott B, Shen J, Nizzero S, Boom K, Persano S, Mi Y, Liu X, Zhao Y, Blanco E, Shen H, Ferrari M, Wolfram J. A pyruvate decarboxylase-mediated therapeutic strategy for mimicking yeast metabolism in cancer cells. Pharmacol Res 2016; 111:413-421. [PMID: 27394167 DOI: 10.1016/j.phrs.2016.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/28/2016] [Accepted: 07/05/2016] [Indexed: 01/18/2023]
Abstract
Cancer cells have high rates of glycolysis and lactic acid fermentation in order to fuel accelerated rates of cell division (Warburg effect). Here, we present a strategy for merging cancer and yeast metabolism to remove pyruvate, a key intermediate of cancer cell metabolism, and produce the toxic compound acetaldehyde. This approach was achieved by administering the yeast enzyme pyruvate decarboxylase to triple negative breast cancer cells. To overcome the challenges of protein delivery, a nanoparticle-based system consisting of cationic lipids and porous silicon were employed to obtain efficient intracellular uptake. The results demonstrate that the enzyme therapy decreases cancer cell viability through production of acetaldehyde and reduction of lactic acid fermentation.
Collapse
Affiliation(s)
- Bronwyn Scott
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jianliang Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Sara Nizzero
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Applied Physics Graduate Program, Rice University, Houston, TX 77005, USA
| | - Kathryn Boom
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Stefano Persano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yu Mi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China; Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
41
|
Conde J, Oliva N, Artzi N. Revisiting the 'One Material Fits All' Rule for Cancer Nanotherapy. Trends Biotechnol 2016; 34:618-626. [PMID: 27262508 DOI: 10.1016/j.tibtech.2016.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 01/12/2023]
Abstract
The promise of (nano)biomaterials for the treatment of cancer can only be realized following a comprehensive scrutiny of the tumor microenvironment. The generic use of 'inert' vehicles that deliver a specific cargo to treat a range of cancer types and disease states obeys the 'one material fits all' rule. However, this approach leads to suboptimal and unpredictable clinical outcomes. The key factors constructing the tumor milieu should guide the design of disease-responsive materials. Given the growing availability of nanomaterials for cancer therapy, a material that responds to each patient's needs and, hence, reacts in a graded manner based on disease cues, would pave the way to precision materials for cancer therapy.
Collapse
Affiliation(s)
- João Conde
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Cambridge, MA, USA; School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| | - Nuria Oliva
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Cambridge, MA, USA
| | - Natalie Artzi
- Massachusetts Institute of Technology, Institute for Medical Engineering and Science, Cambridge, MA, USA; Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|