1
|
Wu M, Li J, Xu Y, Lv S, Wang G, Huang Y, Tao Y, Ju C, Zhang C. Sequentially conquering transfection obstacles of primary human T cells via non-classical lipid nanoparticles delivering reformative transposon system. J Control Release 2025; 383:113779. [PMID: 40306577 DOI: 10.1016/j.jconrel.2025.113779] [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: 12/04/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Chimeric antigen receptor (CAR)-T cells have shown promising efficacy in disease treatment. Considering the safety issues of viral vectors, non-viral vectors such as lipid nanoparticles (LNPs) or transposon systems have attracted growing attention in constructing CAR-T cells. However, the limited transfection efficiency of LNPs and the possible cellular damage via electroporation of transposon system restrict their applications. Herein, we propose to construct CAR-T cells with long-lasting CAR expression via non-classical LNPs delivering reformative piggyBac transposon system. The non-classical LNPs (TA7AD8 LNPs) significantly enhance the cellular uptake by T cells and promote lysosomal escape due to the engagement of novel lipid (AD8) holding asymmetric tails. To further improve the transfection efficiency of transposon, we modify the transposon through inserting a nuclear-targeting sequence and reducing the molecular size to gain a minicircle nuclear location piggyBac transposon system. Based on these, we successfully construct the CD19 CAR-T with long-lasting CAR expression and potent cytotoxicity, achieving antitumor efficacy comparable to that by lentivirus. This work provides a safe, facile, and effective approach for in vitro engineering CAR-T cells, which can be applied in the future to avoid the safety concerns of viral vectors, lower the price, and shorten the manufacturing period of CAR-T cells.
Collapse
Affiliation(s)
- Mengtong Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jingwen Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yazhou Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Shaoxiong Lv
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Guan Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yingshuang Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Tao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
2
|
Xu J, Liu W, Fan F, Zhang B, Sun C, Hu Y. Advances in nano-immunotherapy for hematological malignancies. Exp Hematol Oncol 2024; 13:57. [PMID: 38796455 PMCID: PMC11128130 DOI: 10.1186/s40164-024-00525-3] [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: 11/25/2023] [Accepted: 05/18/2024] [Indexed: 05/28/2024] Open
Abstract
Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Jian Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wenqi Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Fengjuan Fan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
3
|
Zhao X, Amevor FK, Xue X, Wang C, Cui Z, Dai S, Peng C, Li Y. Remodeling the hepatic fibrotic microenvironment with emerging nanotherapeutics: a comprehensive review. J Nanobiotechnology 2023; 21:121. [PMID: 37029392 PMCID: PMC10081370 DOI: 10.1186/s12951-023-01876-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/30/2023] [Indexed: 04/09/2023] Open
Abstract
Liver fibrosis could be the last hope for treating liver cancer and remodeling of the hepatic microenvironment has emerged as a strategy to promote the ablation of liver fibrosis. In recent years, especially with the rapid development of nanomedicine, hepatic microenvironment therapy has been widely researched in studies concerning liver cancer and fibrosis. In this comprehensive review, we summarized recent advances in nano therapy-based remodeling of the hepatic microenvironment. Firstly, we discussed novel strategies for regulatory immune suppression caused by capillarization of liver sinusoidal endothelial cells (LSECs) and macrophage polarization. Furthermore, metabolic reprogramming and extracellular matrix (ECM) deposition are caused by the activation of hepatic stellate cells (HSCs). In addition, recent advances in ROS, hypoxia, and impaired vascular remodeling in the hepatic fibrotic microenvironment due to ECM deposition have also been summarized. Finally, emerging nanotherapeutic approaches based on correlated signals were discussed in this review. We have proposed novel strategies such as engineered nanotherapeutics targeting antigen-presenting cells (APCs) or direct targeting T cells in liver fibrotic immunotherapy to be used in preventing liver fibrosis. In summary, this comprehensive review illustrated the opportunities in drug targeting and nanomedicine, and the current challenges to be addressed.
Collapse
Affiliation(s)
- Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing, 400715, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, 611137, China.
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- , No. 1166, Liu Tai Avenue, Wenjiang district, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Haist M, Mailänder V, Bros M. Nanodrugs Targeting T Cells in Tumor Therapy. Front Immunol 2022; 13:912594. [PMID: 35693776 PMCID: PMC9174908 DOI: 10.3389/fimmu.2022.912594] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
In contrast to conventional anti-tumor agents, nano-carriers allow co-delivery of distinct drugs in a cell type-specific manner. So far, many nanodrug-based immunotherapeutic approaches aim to target and kill tumor cells directly or to address antigen presenting cells (APC) like dendritic cells (DC) in order to elicit tumor antigen-specific T cell responses. Regulatory T cells (Treg) constitute a major obstacle in tumor therapy by inducing a pro-tolerogenic state in APC and inhibiting T cell activation and T effector cell activity. This review aims to summarize nanodrug-based strategies that aim to address and reprogram Treg to overcome their immunomodulatory activity and to revert the exhaustive state of T effector cells. Further, we will also discuss nano-carrier-based approaches to introduce tumor antigen-specific chimeric antigen receptors (CAR) into T cells for CAR-T cell therapy which constitutes a complementary approach to DC-focused vaccination.
Collapse
Affiliation(s)
| | | | - Matthias Bros
- University Medical Center Mainz, Department of Dermatology, Mainz, Germany
| |
Collapse
|
5
|
Boosz P, Pfister F, Stein R, Friedrich B, Fester L, Band J, Mühlberger M, Schreiber E, Lyer S, Dudziak D, Alexiou C, Janko C. Citrate-Coated Superparamagnetic Iron Oxide Nanoparticles Enable a Stable Non-Spilling Loading of T Cells and Their Magnetic Accumulation. Cancers (Basel) 2021; 13:4143. [PMID: 34439296 PMCID: PMC8394404 DOI: 10.3390/cancers13164143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023] Open
Abstract
T cell infiltration into a tumor is associated with a good clinical prognosis of the patient and adoptive T cell therapy can increase anti-tumor immune responses. However, immune cells are often excluded from tumor infiltration and can lack activation due to the immune-suppressive tumor microenvironment. To make T cells controllable by external forces, we loaded primary human CD3+ T cells with citrate-coated superparamagnetic iron oxide nanoparticles (SPIONs). Since the efficacy of magnetic targeting depends on the amount of SPION loading, we investigated how experimental conditions influence nanoparticle uptake and viability of cells. We found that loading in the presence of serum improved both the colloidal stability of SPIONs and viability of T cells, whereas stimulation with CD3/CD28/CD2 and IL-2 did not influence nanoparticle uptake. Furthermore, SPION loading did not impair cytokine secretion after polyclonal stimulation. We finally achieved 1.4 pg iron loading per cell, which was both located intracellularly in vesicles and bound to the plasma membrane. Importantly, nanoparticles did not spill over to non-loaded cells. Since SPION-loading enabled efficient magnetic accumulation of T cells in vitro under dynamic conditions, we conclude that this might be a good starting point for the investigation of in vivo delivery of immune cells.
Collapse
Affiliation(s)
- Philipp Boosz
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Felix Pfister
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Rene Stein
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Lars Fester
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Julia Band
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Marina Mühlberger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Eveline Schreiber
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Medical Immunology Campus Erlangen, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (P.B.); (F.P.); (R.S.); (B.F.); (J.B.); (M.M.); (E.S.); (S.L.); (C.A.)
| |
Collapse
|
6
|
Friedrich RP, Janko C, Unterweger H, Lyer S, Alexiou C. SPIONs and magnetic hybrid materials: Synthesis, toxicology and biomedical applications. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2019-0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract
In the past decades, a wide variety of different superparamagnetic iron oxide nanoparticles (SPIONs) have been synthesized. Due to their unique properties, such as big surface-to-volume ratio, superparamagnetism and comparatively low toxicity, they are principally well suited for many different technical and biomedical applications. Meanwhile, there are a numerous synthesis methods for SPIONs, but high requirements for biocompatibility have so far delayed a successful translation into the clinic. Moreover, depending on the planned application, such as for imaging, magnetic drug targeting, hyperthermia or for hybrid materials intended for regenerative medicine, specific physicochemical and biological properties are inevitable. Since a summary of all existing SPION systems, their properties and application is far too extensive, this review reports on selected methods for SPION synthesis, their biocompatibility and biomedical applications.
Collapse
Affiliation(s)
- Ralf P. Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| |
Collapse
|
7
|
Zheng C, Zhang J, Chan HF, Hu H, Lv S, Na N, Tao Y, Li M. Engineering Nano-Therapeutics to Boost Adoptive Cell Therapy for Cancer Treatment. SMALL METHODS 2021; 5:e2001191. [PMID: 34928094 DOI: 10.1002/smtd.202001191] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/22/2021] [Indexed: 06/14/2023]
Abstract
Although adoptive transfer of therapeutic cells to cancer patients is demonstrated with great success and fortunately approved for the treatment of leukemia and B-cell lymphoma, potential issues, including the unclear mechanism, complicated procedures, unfavorable therapeutic efficacy for solid tumors, and side effects, still hinder its extensive applications. The explosion of nanotechnology recently has led to advanced development of novel strategies to address these challenges, facilitating the design of nano-therapeutics to improve adoptive cell therapy (ACT) for cancer treatment. In this review, the emerging nano-enabled approaches, that design multiscale artificial antigen-presenting cells for cell proliferation and stimulation in vitro, promote the transducing efficiency of tumor-targeting domains, engineer therapeutic cells for in vivo imaging, tumor infiltration, and in vivo functional sustainability, as well as generate tumoricidal T cells in vivo, are summarized. Meanwhile, the current challenges and future perspectives of the nanostrategy-based ACT for cancer treatment are also discussed in the end.
Collapse
Affiliation(s)
- Chunxiong Zheng
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Shixian Lv
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou, 510630, China
| |
Collapse
|
8
|
Cevaal PM, Ali A, Czuba-Wojnilowicz E, Symons J, Lewin SR, Cortez-Jugo C, Caruso F. In Vivo T Cell-Targeting Nanoparticle Drug Delivery Systems: Considerations for Rational Design. ACS NANO 2021; 15:3736-3753. [PMID: 33600163 DOI: 10.1021/acsnano.0c09514] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells play an important role in immunity and repair and are implicated in diseases, including blood cancers, viral infections, and inflammation, making them attractive targets for the treatment and prevention of diseases. Over recent years, the advent of nanomedicine has shown an increase in studies that use nanoparticles as carriers to deliver therapeutic cargo to T cells for ex vivo and in vivo applications. Nanoparticle-based delivery has several advantages, including the ability to load and protect a variety of drugs, control drug release, improve drug pharmacokinetics and biodistribution, and site- or cell-specific targeting. However, the delivery of nanoparticles to T cells remains a major technological challenge, which is primarily due to the nonphagocytic nature of T cells. In this review, we discuss the physiological barriers to effective T cell targeting and describe the different approaches used to deliver cargo-loaded nanoparticles to T cells for the treatment of disease such as T cell lymphoma and human immunodeficiency virus (HIV). In particular, engineering strategies that aim to improve nanoparticle internalization by T cells, including ligand-based targeting, will be highlighted. These nanoparticle engineering approaches are expected to inspire the development of effective nanomaterials that can target or manipulate the function of T cells for the treatment of T cell-related diseases.
Collapse
Affiliation(s)
| | | | - Ewa Czuba-Wojnilowicz
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | - Sharon R Lewin
- Victorian Infectious Diseases, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia
| | - Christina Cortez-Jugo
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
9
|
Sanz-Ortega L, Rojas JM, Barber DF. Improving Tumor Retention of Effector Cells in Adoptive Cell Transfer Therapies by Magnetic Targeting. Pharmaceutics 2020; 12:E812. [PMID: 32867162 PMCID: PMC7557387 DOI: 10.3390/pharmaceutics12090812] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Adoptive cell transfer therapy is a promising anti-tumor immunotherapy in which effector immune cells are transferred to patients to treat tumors. However, one of its main limitations is the inefficient trafficking of inoculated effector cells to the tumor site and the small percentage of effector cells that remain activated when reaching the tumor. Multiple strategies have been attempted to improve the entry of effector cells into the tumor environment, often based on tumor types. It would be, however, interesting to develop a more general approach, to improve and facilitate the migration of specific activated effector lymphoid cells to any tumor type. We and others have recently demonstrated the potential for adoptive cell transfer therapy of the combined use of magnetic nanoparticle-loaded lymphoid effector cells together with the application of an external magnetic field to promote the accumulation and retention of lymphoid cells in specific body locations. The aim of this review is to summarize and highlight the recent findings in the field of magnetic accumulation and retention of effector cells in tumors after adoptive transfer, and to discuss the possibility of using this approach for tumor targeting with chimeric antigen receptor (CAR) T-cells.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine, Karolinska Institute, 14183 Stockholm, Sweden;
| | - José Manuel Rojas
- Animal Health Research Centre (CISA)-INIA, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28130 Madrid, Spain;
| | - Domingo F. Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, 28049 Madrid, Spain
| |
Collapse
|
10
|
Nawaz W, Xu S, Li Y, Huang B, Wu X, Wu Z. Nanotechnology and immunoengineering: How nanotechnology can boost CAR-T therapy. Acta Biomater 2020; 109:21-36. [PMID: 32294554 DOI: 10.1016/j.actbio.2020.04.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/29/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) therapy has achieved remarkable clinical efficacy against hematological cancers and has been approved by FDA for treatment of B-cell tumors. However, the complex manufacturing process and limited success in solid tumors hamper its widespread applications, thus prompting the development of new strategies for overcoming the abovementioned hurdles. In the last decade, nanotechnology has provided sustainable strategies for improving cancer immunotherapy through vaccine development and delivery of immunomodulatory drugs. Nanotechnology can boost CAR-T therapy and may overcome the existing challenges by emerging as a carrier for CAR-T therapy or in combination with CAR-T, it may inhibit solid tumors more effectively than conventional approaches. The revealing of cellular mechanisms, barriers and potential strategies that could be used to manipulate and/or modify cells would enable unprecedented advances in nanotechnology for biologics delivery. This review outlines the journey and barriers of nanoparticles (NPs) across the cell. Subsequently, the approaches to tackle the barriers and strategies to modulate NPs as a carrier for CAR-T therapy are discussed. Finally, the role of NPs in CAR-T therapy and the potential challenges are summarized. This review aims to provide the readers with a detailed overview of NP-based CAR-T therapy research and distil this information into an accessible form conducive to design desired CAR-T therapy using NP approach. STATEMENT OF SIGNIFICANCE: Chimeric antigen receptor (CAR) T-cell therapy is the most vibrant field in immuno-oncology today, with enormous benefits to patients with B-cell malignancies. However, a rapid and straightforward procedure for CAR-T generation is an exigent need to broaden its therapeutic avenue. Nanotechnology has emerged as a novel alternative approach for CAR-T generation. To the best of our knowledge, this is the first in-depth review that briefly highlights the various aspects of nanotechnology in CAR-T therapy, including the strategies to brand NPs as an effective carrier for CAR cargo, its potential advantages, challenges, and future roadmap. It provides readers with a detailed overview of NP-based CAR-T therapy research, and researchers would be able to distill this information into an accessible form conducive to design the desired CAR therapy using the nanotechnology approach.
Collapse
|
11
|
Lee SWL, Paoletti C, Campisi M, Osaki T, Adriani G, Kamm RD, Mattu C, Chiono V. MicroRNA delivery through nanoparticles. J Control Release 2019; 313:80-95. [PMID: 31622695 PMCID: PMC6900258 DOI: 10.1016/j.jconrel.2019.10.007] [Citation(s) in RCA: 261] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are attracting a growing interest in the scientific community due to their central role in the etiology of major diseases. On the other hand, nanoparticle carriers offer unprecedented opportunities for cell specific controlled delivery of miRNAs for therapeutic purposes. This review critically discusses the use of nanoparticles for the delivery of miRNA-based therapeutics in the treatment of cancer and neurodegenerative disorders and for tissue regeneration. A fresh perspective is presented on the design and characterization of nanocarriers to accelerate translation from basic research to clinical application of miRNA-nanoparticles. Main challenges in the engineering of miRNA-loaded nanoparticles are discussed, and key application examples are highlighted to underline their therapeutic potential for effective and personalized medicine.
Collapse
Affiliation(s)
- Sharon Wei Ling Lee
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy; Singapore-MIT Alliance for Research & Technology (SMART), BioSystems and Micromechanics (BioSyM), Singapore, Singapore(3); Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore(3); Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore, Singapore(3)
| | - Camilla Paoletti
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| | - Marco Campisi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| | - Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA; Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo 153-8505, Japan(3)
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore, Singapore(3); Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research & Technology (SMART), BioSystems and Micromechanics (BioSyM), Singapore, Singapore(3); Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy.
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
12
|
Sanz-Ortega L, Rojas JM, Portilla Y, Pérez-Yagüe S, Barber DF. Magnetic Nanoparticles Attached to the NK Cell Surface for Tumor Targeting in Adoptive Transfer Therapies Does Not Affect Cellular Effector Functions. Front Immunol 2019; 10:2073. [PMID: 31543880 PMCID: PMC6728794 DOI: 10.3389/fimmu.2019.02073] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/16/2019] [Indexed: 02/05/2023] Open
Abstract
Adoptive cell transfer therapy is currently one of the most promising approaches for cancer treatment. This therapy has some limitations, however, such as the dispersion of in vivo-administered cells, causing only a small proportion to reach the tumor. Nanotechnological approaches could offer a solution for this drawback, as they can increase cell retention and accumulation in a region of interest. In particular, strategies employing magnetic nanoparticles (MNPs) to improve targeting of adoptively transferred T or NK cells have been explored in mice. In vivo magnetic retention is reported using the human NK cell line NK-92MI transfected with MNPs. Primary NK cells are nonetheless highly resistant to transfection, and thus we explore in here the possibility of attaching the MNPs to the NK cell surface to overcome this issue, and examine whether this association would affect NK effector functions. We assessed the attachment of MNPs coated with different polymers to the NK cell surface, and found that APS-MNP attached more efficiently to the NK-92MI cell surface. In association with MNPs, these cells preserved their main functions, exhibiting a continued capacity to degranulate, conjugate with and lyse target cells, produce IFN-γ, and respond to chemotactic signals. MNP-loaded NK-92MI cells were also retained in an in vitro capillary flow system by applying an EMF. A similar analysis was carried out in primary NK cells, isolated from mice, and expanded in vitro. These primary murine NK cells also maintained their functionality intact after MNP treatment and were successfully retained in vitro. This work therefore provides further support for using MNPs in combination with EMFs to favor specific retention of functional NK cells in a region of interest, which may prove beneficial to adoptive cell-therapy protocols.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - José M Rojas
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| |
Collapse
|
13
|
Sanz-Ortega L, Portilla Y, Pérez-Yagüe S, Barber DF. Magnetic targeting of adoptively transferred tumour-specific nanoparticle-loaded CD8 + T cells does not improve their tumour infiltration in a mouse model of cancer but promotes the retention of these cells in tumour-draining lymph nodes. J Nanobiotechnology 2019; 17:87. [PMID: 31387604 PMCID: PMC6683429 DOI: 10.1186/s12951-019-0520-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background Adoptive T cell-transfer (ATC) therapy is a highly promising cancer-treatment approach. However, in vivo-administered T cells tend to disperse, with only a small proportion reaching the tumour. To remedy this, magnetic targeting of T cells has been recently explored. Magnetic nanoparticles (MNPs) functionalised with antibodies were attached to effector T cells and magnetically recruited to tumour sites under MRI guidance. In this study, we investigated whether 3-aminopropyl-triethoxysilane (APS)-coated MNPs directly attached to CD8+ T cell membranes could also magnetically target and accumulate tumour-specific CD8+ T cells in solid tumours using an external magnetic field (EMF). As it has been shown that T cells associated with APS-coated MNPs are retained in lymph nodes (LNs), and tumour-draining LNs are the most common sites of solid-tumour metastases, we further evaluated whether magnetic targeting of APS-MNP-loaded CD8+ T cells could cause them to accumulate in tumour-draining LNs. Results First, we show that antigen-specific CD8+ T cells preserve their antitumor activity in vitro when associated with APS-MNPs. Next, we demonstrate that the application of a magnetic field enhanced the retention of APS-MNP-loaded OT-I CD8+ T cells under flow conditions in vitro. Using a syngeneic mouse model, we found similar numbers of APS-MNP-loaded OT-I CD8+ T cells and OT-I CD8+ T cells infiltrating the tumour 14 days after cell transfer. However, when a magnet was placed near the tumour during the transfer of tumour-specific APS-MNP-loaded CD8+ T cells to improve tumour infiltration, a reduced percentage of tumour-specific T cells was found infiltrating the tumour 14 days after cell transfer, which was reflected in a smaller reduction in tumour size compared to tumour-specific CD8+ T cells transferred with or without MNPs in the absence of a magnetic field. Nonetheless, magnet placement near the tumour site during cell transfer induced infiltration of activated tumour-specific CD8+ T cells in tumour-draining LNs, which remained 14 days after cell transfer. Conclusions The use of an EMF to improve targeting of tumour-specific T cells modified with APS-MNPs reduced the percentage of these cells infiltrating the tumour, but promoted the retention and the persistence of these cells in the tumour-draining LNs. ![]() Electronic supplementary material The online version of this article (10.1186/s12951-019-0520-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Sanz-Ortega
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology, and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB)-CSIC, Darwin 3, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
14
|
Targeting immune cells for cancer therapy. Redox Biol 2019; 25:101174. [PMID: 30917934 PMCID: PMC6859550 DOI: 10.1016/j.redox.2019.101174] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/08/2019] [Accepted: 03/17/2019] [Indexed: 12/29/2022] Open
Abstract
Recent years have seen a renaissance in the research linking inflammation and cancer with immune cells playing a central role in smouldering inflammation in the tumor microenvironment. Diverse immune cell types infiltrate the tumor microenvironment, and the dynamic tumor-immune cell interplay gives rise to a rich milieu of cytokines and growth factors. Fundamentally, this intricate cross-talk creates the conducive condition for tumor cell proliferation, survival and metastasis. Interestingly, the prominent impact of immune cells is expounded in their contrary pro-tumoral role, as well as their potential anti-cancer cellular weaponry. The latter is known as immunotherapy, a concept born out of evidence that tumors are susceptible to immune defence and that by manipulating the immune system, tumor growth can be successfully restrained. Naturally, a deeper understanding of the multifaceted roles of various immune cell types thus contributes toward developing innovative anti-cancer strategies. Therefore, in this review we first outline the roles played by the major immune cell types, such as macrophages, neutrophils, natural killer cells, T cells and B cells. We then explain the recently-explored strategies of immunomodulation and discuss some important approaches via an immunology perspective.
Collapse
|
15
|
Easo SL, Mohanan PV. Toxicological evaluation of dextran stabilized iron oxide nanoparticles in human peripheral blood lymphocytes. Biointerphases 2016; 11:04B302. [PMID: 27629807 PMCID: PMC5035300 DOI: 10.1116/1.4962268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/20/2016] [Accepted: 08/24/2016] [Indexed: 01/15/2023] Open
Abstract
Iron oxide nanoparticles present an attractive choice for carcinogenic cell destruction via hyperthermia treatment due to its small size and magnetic susceptibility. Dextran stabilized iron oxide nanoparticles (DIONPs) synthesized and characterized for this purpose were used to evaluate its effect on cellular uptake, cytotoxicity, and oxidative stress response in human peripheral blood lymphocytes. In the absence of efficient internalization and perceptible apoptosis, DIONPs were still capable of inducing significant levels of reactive oxygen species formation shortly after exposure. Although these particles did not cause any genotoxic effect, they enhanced the expression of a few relevant oxidative stress and antioxidant defense related genes, accompanied by an increase in the glutathione peroxidase activity. These results indicate that under the tested conditions, DIONPs induced only minimal levels of oxidative stress in lymphocytes. Understanding the biological interaction of DIONPs, the consequences as well as the associated mechanisms in vitro, together with information obtained from systemic studies, could be expected to advance the use of these particles for further clinical trials.
Collapse
Affiliation(s)
- Sheeja Liza Easo
- Division of Toxicology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojapura, Thiruvananthapuram 695 012, Kerala, India
| | - Parayanthala Valappil Mohanan
- Division of Toxicology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojapura, Thiruvananthapuram 695 012, Kerala, India
| |
Collapse
|
16
|
Gonzalez L, Kirsch-Volders M. Reprint of “Biomonitoring of genotoxic effects for human exposure to nanomaterials: The challenge ahead”. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:204-216. [DOI: 10.1016/j.mrrev.2016.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/15/2016] [Accepted: 03/01/2016] [Indexed: 12/25/2022]
|
17
|
Jonuleit H, Bopp T, Becker C. Treg cells as potential cellular targets for functionalized nanoparticles in cancer therapy. Nanomedicine (Lond) 2016; 11:2699-2709. [PMID: 27654070 DOI: 10.2217/nnm-2016-0197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Treg cell-mediated immune suppression appears to represent a significant barrier to effective anticancer immune responses and their inactivation or removal is viewed as a potential therapeutic approach. Although suitable tools for selective Treg cell manipulation in man are missing, their number and function can be altered by a number of drugs and biologicals and by reprogramming tumor-infiltrating antigen presenting cells. Nanoparticles offer exceptional new options in drug and gene delivery by prolonging the circulation time of their cargo, protecting it from degradation and promoting its local accumulation in cells and tissues. In tumor therapy, the use of nanoparticles is expected to overcome limitations in drug delivery and provide novel means for cell-specific functional alteration. In this perspective, we summarize strategies suitable for interference with Treg-mediated suppression, discuss the potential use of nanoparticles for this purpose and identify additional, unexplored opportunities.
Collapse
Affiliation(s)
- Helmut Jonuleit
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
18
|
Biomonitoring of genotoxic effects for human exposure to nanomaterials: The challenge ahead. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 768:14-26. [DOI: 10.1016/j.mrrev.2016.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/15/2016] [Accepted: 03/01/2016] [Indexed: 11/19/2022]
|