1
|
Zhou Z, Chen Y, Ba Y, Xu H, Zuo A, Liu S, Zhang Y, Weng S, Ren Y, Luo P, Cheng Q, Zuo L, Zhu S, Zhou X, Zhang C, Chen Y, Han X, Pan T, Liu Z. Revolutionising Cancer Immunotherapy: Advancements and Prospects in Non-Viral CAR-NK Cell Engineering. Cell Prolif 2024:e13791. [PMID: 39731215 DOI: 10.1111/cpr.13791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/14/2024] [Accepted: 11/28/2024] [Indexed: 12/29/2024] Open
Abstract
The recent advancements in cancer immunotherapy have spotlighted the potential of natural killer (NK) cells, particularly chimeric antigen receptor (CAR)-transduced NK cells. These cells, pivotal in innate immunity, offer a rapid and potent response against cancer cells and pathogens without the need for prior sensitization or recognition of peptide antigens. Although NK cell genetic modification is evolving, the viral transduction method continues to be inefficient and fraught with risks, often resulting in cytotoxic outcomes and the possibility of insertional mutagenesis. Consequently, there has been a surge in the development of non-viral transfection technologies to overcome these challenges in NK cell engineering. Non-viral approaches for CAR-NK cell generation are becoming increasingly essential. Cutting-edge techniques such as trogocytosis, electroporation, lipid nanoparticle (LNP) delivery, clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) gene editing and transposons not only enhance the efficiency and safety of CAR-NK cell engineering but also open new avenues for novel therapeutic possibilities. Additionally, the infusion of technologies already successful in CAR T-cell therapy into the CAR-NK paradigm holds immense potential for further advancements. In this review, we present an overview of the potential of NK cells in cancer immunotherapies, as well as non-viral transfection technologies for engineering NK cells.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yifeng Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lulu Zuo
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yukang Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Li X, Omonova Tuychi Qizi C, Mohamed Khamis A, Zhang C, Su Z. Nanotechnology for Enhanced Cytoplasmic and Organelle Delivery of Bioactive Molecules to Immune Cells. Pharm Res 2022; 39:1065-1083. [PMID: 35661086 DOI: 10.1007/s11095-022-03284-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Immune cells stand as a critical component of the immune system to maintain the internal environment homeostasis. The dysfunction of immune cells can result in various life-threatening diseases, including refractory infection, diabetes, cardiovascular disease, and cancer. Therefore, strategies to standardize or even enhance the function of immune cells are critical. Recently, nanotechnology has been highly researched and extensively applied for enhancing the cytoplasmic delivery of bioactive molecules to immune cells, providing efficient approaches to correct in vivo and in vitro dysfunction of immune cells. This review focuses on the technologies and challenges involved in improving endo-lysosomal escape, cytoplasmic release and organelle targeted delivery of different bioactive molecules in immune cells. Furthermore, it will elaborate on the broader vision of applying nanotechnology for treating immune cell-related diseases and constructing immune therapies and cytopharmaceuticals as potential treatments for diseases.
Collapse
Affiliation(s)
- Xiaoyu Li
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Charos Omonova Tuychi Qizi
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Amari Mohamed Khamis
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Center of Advanced Pharmaceuticals and Biomaterials, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
3
|
Potential of cell-penetrating peptides (CPPs) in delivery of antiviral therapeutics and vaccines. Eur J Pharm Sci 2021; 169:106094. [PMID: 34896590 DOI: 10.1016/j.ejps.2021.106094] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Viral infections are a great threat to human health. Currently, there are no effective vaccines and antiviral drugs against the majority of viral diseases, suggesting the need to develop novel and effective antiviral agents. Since the intracellular delivery of antiviral agents, particularly the impermeable molecules, such as peptides, proteins, and nucleic acids, are essential to exert their therapeutic effects, using a delivery system is highly required. Among various delivery systems, cell-penetrating peptides (CPPs), a group of short peptides with the unique ability of crossing cell membrane, offer great potential for the intracellular delivery of various biologically active cargoes. The results of numerous in vitro and in vivo studies with CPP conjugates demonstrate their promise as therapeutic agents in various medical fields including antiviral therapy. The CPP-mediated delivery of various antiviral agents including peptides, proteins, nucleic acids, and nanocarriers have been associated with therapeutic efficacy both in vitro and in vivo. This review describes various aspects of viruses including their biology, pathogenesis, and therapy and briefly discusses the concept of CPP and its potential in drug delivery. Particularly, it will highlight a variety of CPP applications in the management of viral infections.
Collapse
|
4
|
Kunte N, McGraw E, Bell S, Held D, Avila LA. Prospects, challenges and current status of RNAi through insect feeding. PEST MANAGEMENT SCIENCE 2020; 76:26-41. [PMID: 31419022 DOI: 10.1002/ps.5588] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/21/2019] [Accepted: 08/13/2019] [Indexed: 05/06/2023]
Abstract
RNA interference is a phenomenon in which the introduction of double-stranded RNA (dsRNA) into cells triggers the degradation of the complementary messenger RNA in a sequence-specific manner. Suppressing expression of vital genes could lead to insect death, therefore this technology has been considered as a potential strategy for insect pest control. There are three main routes of dsRNA administration into insects: (i) injections to the hemolymph, (ii) topical, and (iii) feeding. In this review, we focus on dsRNA administration through feeding. We summarize novel strategies that have been developed to improve the efficacy of this method, such as the use of nano-based formulations, engineered microorganisms, and transgenic plants. We also expose the hurdles that have to be overcome in order to use this technique as a reliable pest management method. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Nitish Kunte
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | - Erin McGraw
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | - Sydney Bell
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| | - David Held
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL, USA
| | - Luz-Adriana Avila
- Department of Biological Sciences, Auburn University, Auburn, AL, USA
| |
Collapse
|
5
|
Han P, Hanlon D, Sobolev O, Chaudhury R, Edelson RL. Ex vivo dendritic cell generation-A critical comparison of current approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:251-307. [PMID: 31759433 DOI: 10.1016/bs.ircmb.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, required for the initiation of naïve and memory T cell responses and regulation of adaptive immunity. The discovery of DCs in 1973, which culminated in the Nobel Prize in Physiology or Medicine in 2011 for Ralph Steinman and colleagues, initially focused on the identification of adherent mononuclear cell fractions with uniquely stellate dendritic morphology, followed by key discoveries of their critical immunologic role in initiating and maintaining antigen-specific immunity and tolerance. The medical promise of marshaling these key capabilities of DCs for therapeutic modulation of antigen-specific immune responses has guided decades of research in hopes to achieve genuine physiologic partnership with the immune system. The potential uses of DCs in immunotherapeutic applications include cancer, infectious diseases, and autoimmune disorders; thus, methods for rapid and reliable large-scale production of DCs have been of great academic and clinical interest. However, difficulties in obtaining DCs from lymphoid and peripheral tissues, low numbers and poor survival in culture, have led to advancements in ex vivo production of DCs, both for probing molecular details of DC function as well as for experimenting with their clinical utility. Here, we review the development of a diverse array of DC production methodologies, ranging from cytokine-based strategies to genetic engineering tools devised for enhancing DC-specific immunologic functions. Further, we explore the current state of DC therapies in clinic, as well as emerging insights into physiologic production of DCs inspired by existing therapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Rabib Chaudhury
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Richard L Edelson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
6
|
Soler Besumbes E, Fornaguera C, Monge M, García-Celma MJ, Carrión J, Solans C, Dols-Perez A. PLGA cationic nanoparticles, obtained from nano-emulsion templating, as potential DNA vaccines. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.109229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
7
|
Lou B, De Koker S, Lau CYJ, Hennink WE, Mastrobattista E. mRNA Polyplexes with Post-Conjugated GALA Peptides Efficiently Target, Transfect, and Activate Antigen Presenting Cells. Bioconjug Chem 2018; 30:461-475. [PMID: 30188694 PMCID: PMC6385079 DOI: 10.1021/acs.bioconjchem.8b00524] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Vaccines based on
mRNA have emerged as potent systems to elicit
CD8+ T cell responses against various cancers and viral
infectious diseases. The efficient intracellular delivery of mRNA
molecules encoding antigens into the cytosol of antigen-presenting
cells (APCs) is still challenging, requiring cell attachment, active
uptake, and subsequent endosomal escape. Here, we report a facile
approach for the formulation of peptide-functionalized mRNA polyplexes
using copper-free click chemistry to promote presentation of mRNA
antigen by dendritic cells (DCs). After screening different membrane
active peptides, GALA modified mRNA polyplexes (PPx-GALA) with a size
around 350 nm and with a slightly negative surface charge (−7
mV), exhibited the highest EGFP-mRNA transfection in RAW 246.7 macrophages
(∼36%) and D1 dendritic cells (∼50%) as compared to
polyplexes decorated with melittin or LEDE peptides. Interestingly,
we found that PPx-GALA enters DCs through sialic acid mediated endo/phagocytosis,
which was not influenced by DC maturation. The PPx-GALA formulation
exhibited 18-fold higher cellular uptake compared to a lipofectamine
mRNA formulation without inducing cytotoxicity. Live cell imaging
showed that PPx-GALA that were taken up by endocytosis induced calcein
release from endosomes into the cytosol. DCs treated with PPx-GALA
containing mRNA encoding for OVA displayed enhanced T cell responses
and DC maturation. Collectively, these data provide a strong rationale
for further study of this PPx-GALA formulation in vivo as a promising mRNA vaccine platform.
Collapse
Affiliation(s)
- Bo Lou
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Utrecht University , 3584CG Utrecht , The Netherlands
| | - Stefaan De Koker
- Laboratory of Molecular Immunology, Department of Biomedical Molecular Biology , Ghent University , 9052 Zwijnaarde , Belgium
| | - Chun Yin Jerry Lau
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Utrecht University , 3584CG Utrecht , The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Utrecht University , 3584CG Utrecht , The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS) , Utrecht University , 3584CG Utrecht , The Netherlands
| |
Collapse
|
8
|
Recent advances in applying nanotechnologies for cancer immunotherapy. J Control Release 2018; 288:239-263. [PMID: 30223043 DOI: 10.1016/j.jconrel.2018.09.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aimed at boosting cancer-specific immunoresponses to eradicate tumor cells has evolved as a new treatment modality. Nanoparticles incorporating antigens and immunomodulatory agents can activate immune cells and modulate the tumor microenvironment to enhance anti-tumor immunity. The nanotechnology approach has been demonstrated to be superior to standard formulations in in-vivo settings. In this article, we focus on recent advances made within the last 5 years in nanoparticle-based cancer immunotherapy, including peptide- and nucleic acid-based nanovaccines, nanomedicines containing an immunoadjuvant to activate anti-tumor immunity, nanoparticle delivery of immune checkpoint inhibitors and the combination of the above approaches. Encouraging results and new emerging nanotechnologies in drug delivery promise the continuous growth of this field and ultimately clinical translation of enhanced immunotherapy of cancer.
Collapse
|
9
|
Fornaguera C, Guerra-Rebollo M, Ángel Lázaro M, Castells-Sala C, Meca-Cortés O, Ramos-Pérez V, Cascante A, Rubio N, Blanco J, Borrós S. mRNA Delivery System for Targeting Antigen-Presenting Cells In Vivo. Adv Healthc Mater 2018; 7:e1800335. [PMID: 29923337 DOI: 10.1002/adhm.201800335] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/06/2018] [Indexed: 12/13/2022]
Abstract
The encapsulation of mRNA in nanosystems as gene vaccines for immunotherapy purposes has experienced an exponential increase in recent years. Despite the many advantages envisaged within these approaches, their application in clinical treatments is still limited due to safety issues. These issues can be attributed, in part, to liver accumulation of most of the designed nanosystems and to the inability to transfect immune cells after an intravenous administration. In this context, this study takes advantage of the known versatile properties of the oligopeptide end-modified poly (β-amino esters) (OM-PBAEs) to complex mRNA and form discrete nanoparticles. Importantly, it is demonstrated that the selection of the appropriate end-oligopeptide modifications enables the specific targeting and major transfection of antigen-presenting cells (APC) in vivo, after intravenous administration, thus enabling their use for immunotherapy strategies. Therefore, with this study, it can be confirmed that OM-PBAE are appropriate systems for the design of mRNA-based immunotherapy approaches aimed to in vivo transfect APCs and trigger immune responses to fight either tumors or infectious diseases.
Collapse
Affiliation(s)
- Cristina Fornaguera
- Sagetis Biotech SL; 08017 Barcelona Spain
- Grup d'Enginyeria de Materials (GEMAT); Institut Químic de Sarrià (IQS); Universitat Ramon Llull (URL); 08017 Barcelona Spain
| | - Marta Guerra-Rebollo
- CIBER of Biomaterials; Bioengineering and Nanomedicine (CIBER-BBN); 08034 Barcelona Spain
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC); 08034 Barcelona Spain
| | | | | | - Oscar Meca-Cortés
- CIBER of Biomaterials; Bioengineering and Nanomedicine (CIBER-BBN); 08034 Barcelona Spain
| | - Victor Ramos-Pérez
- Grup d'Enginyeria de Materials (GEMAT); Institut Químic de Sarrià (IQS); Universitat Ramon Llull (URL); 08017 Barcelona Spain
| | - Anna Cascante
- Sagetis Biotech SL; 08017 Barcelona Spain
- Grup d'Enginyeria de Materials (GEMAT); Institut Químic de Sarrià (IQS); Universitat Ramon Llull (URL); 08017 Barcelona Spain
| | - Núria Rubio
- CIBER of Biomaterials; Bioengineering and Nanomedicine (CIBER-BBN); 08034 Barcelona Spain
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC); 08034 Barcelona Spain
| | - Jerónimo Blanco
- CIBER of Biomaterials; Bioengineering and Nanomedicine (CIBER-BBN); 08034 Barcelona Spain
- Institute of Advanced Chemistry of Catalonia (IQAC-CSIC); 08034 Barcelona Spain
| | - Salvador Borrós
- Sagetis Biotech SL; 08017 Barcelona Spain
- Grup d'Enginyeria de Materials (GEMAT); Institut Químic de Sarrià (IQS); Universitat Ramon Llull (URL); 08017 Barcelona Spain
- CIBER of Biomaterials; Bioengineering and Nanomedicine (CIBER-BBN); 08034 Barcelona Spain
| |
Collapse
|
10
|
Yamada A, Mitsueda A, Hasan M, Ueda M, Hama S, Warashina S, Nakamura T, Harashima H, Kogure K. Tri-membrane nanoparticles produced by combining liposome fusion and a novel patchwork of bicelles to overcome endosomal and nuclear membrane barriers to cargo delivery. Biomater Sci 2016; 4:439-47. [DOI: 10.1039/c5bm00327j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tri-membrane PD-MEND deliver cargo by sophisticated membrane fusions.
Collapse
Affiliation(s)
- Asako Yamada
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Asako Mitsueda
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Mahadi Hasan
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Miho Ueda
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Susumu Hama
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Shota Warashina
- Laboratory for Molecular Design of Pharmaceutics
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Takashi Nakamura
- Laboratory for Molecular Design of Pharmaceutics
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics
- Faculty of Pharmaceutical Sciences
- Hokkaido University
- Sapporo
- Japan
| | - Kentaro Kogure
- Department of Biophysical Chemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| |
Collapse
|
11
|
Seth A, Oh DB, Lim YT. Nanomaterials for enhanced immunity as an innovative paradigm in nanomedicine. Nanomedicine (Lond) 2015; 10:959-75. [PMID: 25867860 DOI: 10.2217/nnm.14.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since the advent of nanoparticle technology, novel and versatile properties of nanomaterials have been introduced, which has constantly expanded their applications in therapeutics. Introduction of nanomaterials for immunomodulation has opened up new avenues with tremendous potential. Interesting properties of nanoparticles, such as adjuvanticity, capability to enhance cross-presentation, polyvalent presentation, siRNA delivery for silencing of immunesuppressive gene, targeting and imaging of immune cells have been known to have immense utility in vaccination and immunotherapy. A thorough understanding of the merits associated with nanomaterials is crucial for designing of modular and versatile nanovaccines, for improved immune response. With the emerging prerequisites of vaccination, nanomaterial-based immune stimulation, seems to be capable of taking the field of immunization to a next higher level.
Collapse
Affiliation(s)
- Anushree Seth
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 305-764, South Korea
| | | | | |
Collapse
|
12
|
Field LD, Delehanty JB, Chen Y, Medintz IL. Peptides for specifically targeting nanoparticles to cellular organelles: quo vadis? Acc Chem Res 2015; 48:1380-90. [PMID: 25853734 DOI: 10.1021/ar500449v] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The interfacing of nanomaterials and especially nanoparticles within all aspects of biological research continues to grow at a nearly unabated pace with projected applications focusing on powerful new tools for cellular labeling, imaging, and sensing, theranostic materials, and drug delivery. At the most fundamental level, many of these nanoparticles are meant to target not only very specific cell-types, regardless of whether they are in a culture, tissue, an animal model, or ultimately a patient, but also in many cases a specific subcellular organelle. During this process, these materials will undergo a complex journey that must first find the target cell of interest, then be taken up by those cells across the extracellular membrane, and ultimately localize to a desired subcellular organelle, which may include the nucleus, plasma membrane, endolysosomal system, mitochondria, cytosol, or endoplasmic reticulum. To accomplish these complex tasks in the correct sequence, researchers are increasingly interested in selecting for and exploiting targeting peptides that can impart the requisite capabilities to a given nanoparticle construct. There are also a number of related criteria that need careful consideration for this undertaking centering on the nature and properties of the peptide vector itself, the peptide-nanoparticle conjugate characteristics, and the target cell. Here, we highlight some important issues and key research areas related to this burgeoning field. We begin by providing a brief overview of some criteria for optimal attachment of peptides to nanoparticles, the predominant methods by which nanoparticles enter cells, and some of the peptide sequences that have been utilized to facilitate nanoparticle delivery to cells focusing on those that engender the initial targeting and uptake. Because almost all materials delivered to cells by peptides utilize the endosomal system of vesicular transport and in many cases remain sequestered within the vesicles, we critically evaluate the issue of endosomal escape in the context of some recently reported successes in this regard. Following from this, peptides that have been reported to deliver nanoparticles to specific subcellular compartments are examined with a focus on what they delivered and the putative mechanisms by which they were able to accomplish this. The last section focuses on two areas that are critical to realizing this overall approach in the long term. The first is how to select for peptidyl sequences capable of improved or more specific cellular or subcellular targeting based upon principles commonly associated with drug discovery. The second looks at what has been done to create modular peptides that incorporate multiple desirable functionalities within a single, contiguous sequence. This provides a viable alternative to either the almost insurmountable challenge of finding one sequence capable of all functions or, alternatively, attaching different peptides with different functionalities to the same nanoparticle in different ratios when trying to orchestrate their net effects. Finally, we conclude with a brief perspective on the future evolution and broader impact of this growing area of bionanoscience.
Collapse
Affiliation(s)
- Lauren D. Field
- Center for Bio/Molecular Science and Engineering,
Code 6900, U.S. Naval Research Laboratory, 4555 Overlook Avenue, SW, Washington, D.C. 20375, United States
- Fischell Department of Bioengineering, 2330 Kim Engineering Building, University of Maryland, College Park, Maryland 20742, United States
| | - James B. Delehanty
- Center for Bio/Molecular Science and Engineering,
Code 6900, U.S. Naval Research Laboratory, 4555 Overlook Avenue, SW, Washington, D.C. 20375, United States
| | - YungChia Chen
- Center for Bio/Molecular Science and Engineering,
Code 6900, U.S. Naval Research Laboratory, 4555 Overlook Avenue, SW, Washington, D.C. 20375, United States
- American Society for Engineering Education Washington, D.C. 20036, United States
| | - Igor L. Medintz
- Center for Bio/Molecular Science and Engineering,
Code 6900, U.S. Naval Research Laboratory, 4555 Overlook Avenue, SW, Washington, D.C. 20375, United States
| |
Collapse
|
13
|
Furukawa R, Yamada Y, Kawamura E, Harashima H. Mitochondrial delivery of antisense RNA by MITO-Porter results in mitochondrial RNA knockdown, and has a functional impact on mitochondria. Biomaterials 2015; 57:107-15. [PMID: 25913255 DOI: 10.1016/j.biomaterials.2015.04.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 10/23/2022]
Abstract
Mitochondrial genome-targeting nucleic acids are promising therapeutic candidates for treating mitochondrial diseases. To date, a number of systems for delivering genetic information to the cytosol and the nucleus have been reported, and several successful gene therapies involving gene delivery targeted to the cytosol and the nucleus have been reported. However, much less progress has been made concerning mitochondrial gene delivery systems, and mitochondrial gene therapy has never been achieved. Here, we report on the mitochondrial delivery of an antisense RNA oligonucleotide (ASO) to perform mitochondrial RNA knockdown to regulate mitochondrial function. Mitochondrial delivery of the ASO was achieved using a combination of a MITO-Porter system, which contains mitochondrial fusogenic lipid envelopes for mitochondrial delivery via membrane fusion and D-arm, a mitochondrial import signal of tRNA to the matrix. Mitochondrial delivery of the ASO induces the knockdown of the targeted mitochondria-encoded mRNA and protein, namely cytochrome c oxidase subunit II, a component of the mitochondrial respiratory chain. Furthermore, the mitochondrial membrane potential was depolarized by the down regulation of the respiratory chain as the result of the mitochondrial delivery of ASO. This finding constitutes the first report to demonstrate that the nanocarrier-mediated mitochondrial genome targeting of antisense RNA effects mitochondrial function.
Collapse
Affiliation(s)
- Ryo Furukawa
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Eriko Kawamura
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan.
| |
Collapse
|
14
|
Yamada Y, Tabata M, Yasuzaki Y, Nomura M, Shibata A, Ibayashi Y, Taniguchi Y, Sasaki S, Harashima H. A nanocarrier system for the delivery of nucleic acids targeted to a pancreatic beta cell line. Biomaterials 2014; 35:6430-8. [PMID: 24816283 DOI: 10.1016/j.biomaterials.2014.04.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/05/2014] [Indexed: 11/25/2022]
Abstract
Pancreatic β cells secrete insulin in response to glucose levels and thus are involved in controlling blood glucose levels. A line of pancreatic β cells "MIN6" has been used in studies related to the function of β cells and diabetes therapy. Regulating gene expression in MIN6 cells could accelerate these studies, but an efficient method for the transfection of nucleic acids targeted to MIN6 cells is required. We report here on a liposome-based carrier targeted to pancreatic β cells (Multifunctional envelope-type nano device for pancreatic β cells, β-MEND). We identified a lipid composition for use in preparing the β-MEND, which permits the particles to be efficiently internalized into MIN6, as evidenced by flow cytometry analyses. Intracellular observation by confocal laser scanning microscopy showed that the β-MEND efficiently delivered the oligo nucleic acids to the cytosol of MIN6 cells. Moreover, using a β-MEND encapsulating a 2'-O-Methyl RNA complementary to a microRNA that suppresses insulin secretion, the knockdown of the targeted microRNA and an up-regulation of insulin secretion were observed in MIN6. Thus, the β-MEND holds promise as an efficient system for delivering nucleic acids targeted to MIN6 and can contribute to research and therapy aimed at diabetes.
Collapse
Affiliation(s)
- Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Mai Tabata
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yukari Yasuzaki
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Masatoshi Nomura
- Department of Endocrine and Metabolic Diseases/Diabetes Mellitus, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Atsushi Shibata
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuta Ibayashi
- Department of Endocrine and Metabolic Diseases/Diabetes Mellitus, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yosuke Taniguchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shigeki Sasaki
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
15
|
Mitochondrial targeting functional peptides as potential devices for the mitochondrial delivery of a DF-MITO-Porter. Mitochondrion 2013; 13:610-4. [DOI: 10.1016/j.mito.2013.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/28/2013] [Accepted: 08/28/2013] [Indexed: 11/21/2022]
|
16
|
Yasuzaki Y, Yamada Y, Kanefuji T, Harashima H. Localization of exogenous DNA to mitochondria in skeletal muscle following hydrodynamic limb vein injection. J Control Release 2013; 172:805-11. [PMID: 24100263 DOI: 10.1016/j.jconrel.2013.09.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 09/19/2013] [Accepted: 09/26/2013] [Indexed: 01/16/2023]
Abstract
Mitochondrial genetic disorders are a major cause of mitochondrial diseases. It is therefore likely that mitochondrial gene therapy will be useful for the treatment of such diseases. Here, we report on the possibility of mitochondrial gene delivery in skeletal muscle using hydrodynamic limb vein (HLV) injection. The HLV injection procedure, a useful method for transgene expression in skeletal muscle, involves the rapid injection of a large volume of naked plasmid DNA (pDNA) into the distal vein of a limb. We hypothesized that the technique could be used to deliver pDNA not only to nuclei but also to mitochondria, since cytosolic pDNA that is internalized by the method may be able to overcome mitochondrial membrane. We determined if pDNA could be delivered to myofibrillar mitochondria by HLV injection by PCR analysis. Mitochondrial toxicity assays showed that the HLV injection had no influence on mitochondrial function. These findings indicate that HLV injection promises to be a useful technique for in vivo mitochondrial gene delivery.
Collapse
Affiliation(s)
- Yukari Yasuzaki
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | | | |
Collapse
|
17
|
Shen Y, Fang H, Zhang K, Shrestha R, Wooley KL, Taylor JSA. Efficient protection and transfection of small interfering RNA by cationic shell-crosslinked knedel-like nanoparticles. Nucleic Acid Ther 2013; 23:95-108. [PMID: 23557117 DOI: 10.1089/nat.2012.0390] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite the great potential of small interfering RNA (siRNA) as a therapeutic agent, progress in this area has been hampered by a lack of efficient biocompatible transfection agents. Recently, cationic shell-crosslinked knedel-like nanoparticles (cSCKs) were found to possess lower cytotoxicity and better transfection ability for phosphorothioate ODNs and plasmid DNA than the commonly used cationic lipid-based agent Lipofectamine. To determine the usefulness of cSCKs for siRNA transfection, a small library of cSCKs with varying percentage of primary and tertiary amines was assessed for its ability to bind to siRNA, inhibit siRNA degradation in human serum, and to transfect HeLa and mouse macrophage cell lines. The silencing efficiency in HeLa cells was greatest with the cSCK with 100% primary amines (pa100) as determined by their viability following transfection with cytotoxic and non-cytotoxic siRNAs. cSCK-pa100 showed greater silencing efficiency than Lipofectamine 2000 in the HeLa cells, as well in 293T and human bronchial epithelial (HEK) cells, but was comparable in human bronchial epithelial (BEAS-2B) cells and human mammary epithelial (MCF10a) cells. cSCK-pa100 also showed greater silencing of iNOS expression than Lipofectamine 2000 in a mouse macrophage cell line, and provided greater protection from serum degradation, demonstrating its potential usefulness as an siRNA transfection agent. The siRNA silencing of iNOS at lower concentrations of siRNA could be enhanced by complexation with the fusogenic GALA peptide, which was shown to enhance endosomal escape following uptake.
Collapse
Affiliation(s)
- Yuefei Shen
- Department of Chemistry, Washington University , St. Louis, MO 63130, USA
| | | | | | | | | | | |
Collapse
|
18
|
Kitazoe K, Park YS, Kaji N, Okamoto Y, Tokeshi M, Kogure K, Harashima H, Baba Y. Fabrication of functionalized double-lamellar multifunctional envelope-type nanodevices using a microfluidic chip with a chaotic mixer array. PLoS One 2012; 7:e39057. [PMID: 22723929 PMCID: PMC3377610 DOI: 10.1371/journal.pone.0039057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 05/16/2012] [Indexed: 11/30/2022] Open
Abstract
Multifunctional envelope-type nanodevices (MENDs) are very promising non-viral gene delivery vectors because they are biocompatible and enable programmed packaging of various functional elements into an individual nanostructured liposome. Conventionally MENDs have been fabricated by complicated, labor-intensive, time-consuming bulk batch methods. To avoid these problems in MEND fabrication, we adopted a microfluidic chip with a chaotic mixer array on the floor of its reaction channel. The array was composed of 69 cycles of the staggered chaotic mixer with bas-relief structures. Although the reaction channel had very large Péclet numbers (>10(5)) favorable for laminar flows, its chaotic mixer array led to very small mixing lengths (<1.5 cm) and that allowed homogeneous mixing of MEND precursors in a short time. Using the microfluidic chip, we fabricated a double-lamellar MEND (D-MEND) composed of a condensed plasmid DNA core and a lipid bilayer membrane envelope as well as the D-MEND modified with trans-membrane peptide octaarginine. Our lab-on-a-chip approach was much simpler, faster, and more convenient for fabricating the MENDs, as compared with the conventional bulk batch approaches. Further, the physical properties of the on-chip-fabricated MENDs were comparable to or better than those of the bulk batch-fabricated MENDs. Our fabrication strategy using microfluidic chips with short mixing length reaction channels may provide practical ways for constructing more elegant liposome-based non-viral vectors that can effectively penetrate all membranes in cells and lead to high gene transfection efficiency.
Collapse
Affiliation(s)
- Katsuma Kitazoe
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Yeon-Su Park
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- FIRST Research Center for Innovative Nanobiodevices, Nagoya University, Nagoya, Japan
| | - Noritada Kaji
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- FIRST Research Center for Innovative Nanobiodevices, Nagoya University, Nagoya, Japan
| | - Yukihiro Okamoto
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- FIRST Research Center for Innovative Nanobiodevices, Nagoya University, Nagoya, Japan
| | - Manabu Tokeshi
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- FIRST Research Center for Innovative Nanobiodevices, Nagoya University, Nagoya, Japan
| | - Kentaro Kogure
- Department of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshinobu Baba
- Department of Applied Chemistry, Graduate School of Engineering, Nagoya University, Nagoya, Japan
- FIRST Research Center for Innovative Nanobiodevices, Nagoya University, Nagoya, Japan
- Health Technology Research Center, National Institute of Advanced Industrial Science and Technology, Takamatsu, Japan
| |
Collapse
|
19
|
|
20
|
Abstract
RNA interference (RNAi) has been extensively employed for in vivo research since its use was first demonstrated in mammalian cells 10 years ago. Design rules have improved, and it is now routinely possible to obtain reagents that suppress expression of any gene desired. At the same time, increased understanding of the molecular basis of unwanted side effects has led to the development of chemical modification strategies that mitigate these concerns. Delivery remains the single greatest hurdle to widespread adoption of in vivo RNAi methods. However, exciting advances have been made and new delivery systems under development may help to overcome these barriers. This review discusses advances in RNAi biochemistry and biology that impact in vivo use and provides an overview of select publications that demonstrate interesting applications of these principles. Emphasis is placed on work with synthetic, small interfering RNAs (siRNAs) published since the first installment of this review which appeared in 2006.
Collapse
|