1
|
Syed Altaf RR, Mohan A, Palani N, Mendonce KC, Monisha P, Rajadesingu S. A review of innovative design strategies: Artificial antigen presenting cells in cancer immunotherapy. Int J Pharm 2025; 669:125053. [PMID: 39667594 DOI: 10.1016/j.ijpharm.2024.125053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Developing nanocarriers that can carry medications directly to tumors is an exciting development in cancer nanomedicine. The efficacy of this intriguing therapeutic approach is, however, compromised by intricate and immunosuppressive circumstances that arise concurrently with the onset of cancer. The artificial antigen presenting cell (aAPC), a micro or nanoparticle based device that mimics an antigen presenting cell by providing crucial signal proteins to T lymphocytes to activate them against cancer, is one cutting-edge method for cancer immunotherapy. This review delves into the critical design considerations for aAPCs, particularly focusing on particle size, shape, and the non-uniform distribution of T cell activating proteins on their surfaces. Adequate surface contact between T cells and aAPCs is essential for activation, prompting engineers to develop nano-aAPCs with microscale contact areas through techniques such as shape modification and nanoparticle clustering. Additionally, we explore recommendations for future advancements in this field.
Collapse
Affiliation(s)
- Rabiya Riffath Syed Altaf
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Agilandeswari Mohan
- Department of BioChemistry, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Naveen Palani
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Keren Celestina Mendonce
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India; Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - P Monisha
- PG & Research Department of Physics, Sri Sarada College for Women, Salem - 636016, Tamil Nadu, India
| | - Suriyaprakash Rajadesingu
- Centre for Research in Environment, Sustainability Advocacy and Climate CHange (REACH), Directorate of Research, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
2
|
Hong H, Park CH, Lee JS, Kim K, Nath S, Oh MS, Kim S, Lee CH, Kim KH, Choi WH, Choi KY, Park HS, Lee OJ, Hong IS, Kim SH. Ex vivo enhancement of CD8+ T cell activity using functionalized hydrogel encapsulating tonsil-derived lymphatic endothelial cells. Theranostics 2025; 15:850-874. [PMID: 39776798 PMCID: PMC11700866 DOI: 10.7150/thno.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Rationale: This study investigates a method for programming immune cells using a biomaterial-based system, providing an alternative to traditional ex vivo cell manipulation techniques. It addresses the limitations of engineered adoptive T cell therapies, such as T cell exhaustion, by introducing a gelatin-hyaluronic acid (GH-GMA) hydrogel system. Methods: We characterized tonsil mesenchymal stem cells (TMSCs), lymphatic endothelial cells (T-LECs), stimulated T-CD8+ T cells (STCs), and GH-GMA biomaterials. The 10% 5:1 GH-GMA hydrogel, loaded with anti-CD28, cytokines interleukin-2 (IL-2) and vascular endothelial growth factor C (VEGF-C), forms a functional hydrogel capable of releasing these immune-stimulating factors. T-LEC spheroids, derived from tonsil mesenchymal stem cells (TMSCs), were encapsulated within the hydrogel to act as antigen-presenting cells for T cells. Results: Co-encapsulation of STCs and T-LEC spheroids in the functional hydrogel resulted in significant expansion and enrichment of STCs during cultivation. Moreover, when cancer cells were co-encapsulated with STCs and T-LECs, there was increased migration of STCs towards the cancer cells and elevated expression of PD-L1 on the cancer cells. Conclusions: These findings suggest that the GH-GMA hydrogel, combined with anti-CD28, IL-2, VEGF-C, and T-LEC spheroids, enhances T cell activity, presenting a promising platform for cancer immunotherapies and modulation of the suppressive tumor microenvironment.
Collapse
Affiliation(s)
- Heesun Hong
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hos-pital, School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyunghee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sudarshini Nath
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Moon Sik Oh
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sol Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Chul Hee Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ki Hyun Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Woo Hee Choi
- R&D Institute, ORGANOIDSCIENCES Ltd., Seongnam 13488, Republic of Korea
| | - Kyu Young Choi
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Hae Sang Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hos-pital, School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 21565, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
3
|
Varghese B, González-Navarro JA, Guerra VLP, Faizulina M, Artemieva D, Chum T, Ramakrishna TRB, Cebecauer M, Kovaříček P. Cell surface morphology mimicking nano-bio platform for immune cell stimulation. iScience 2024; 27:111033. [PMID: 39498306 PMCID: PMC11532961 DOI: 10.1016/j.isci.2024.111033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 11/07/2024] Open
Abstract
Studying the complex realm of cellular communication and interactions by fluorescence microscopy requires sample fixation on a transparent substrate. To activate T cells, which are pivotal in controlling the immune system, it is important to present the activating antigen in a spatial arrangement similar to the nature of the antigen-presenting cell, including the presence of ligands on microvilli. Similar arrangement is predicted for some other immune cells. In this work, immune cell-stimulating platform based on nanoparticle-ligand conjugates have been developed using a scalable, affordable, and broadly applicable technology, which can be readily deployed without the need for state-of-the-art nanofabrication instruments. The validation of surface biofunctionalization was performed by combination of fluorescence and atomic force microscopy techniques. We demonstrate that the targeted system serves as a biomimetic scaffold on which immune cells make primary contact with the microvilli-mimicking substrate and exhibit stimulus-specific activation.
Collapse
Affiliation(s)
- Beena Varghese
- Department of Organic Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czechia
| | - José Alfredo González-Navarro
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague, Czechia
| | - Valentino Libero Pio Guerra
- Department of Organic Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czechia
| | - Margarita Faizulina
- Department of Organic Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czechia
| | - Daria Artemieva
- Department of Organic Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czechia
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague, Czechia
| | - Tomáš Chum
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague, Czechia
| | - Tejaswini Rama Bangalore Ramakrishna
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague, Czechia
| | - Marek Cebecauer
- Department of Biophysical Chemistry, J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague, Czechia
| | - Petr Kovaříček
- Department of Organic Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czechia
| |
Collapse
|
4
|
Wells K, Liu T, Zhu L, Yang L. Immunomodulatory nanoparticles activate cytotoxic T cells for enhancement of the effect of cancer immunotherapy. NANOSCALE 2024; 16:17699-17722. [PMID: 39257225 DOI: 10.1039/d4nr01780c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Cancer immunotherapy represents a promising targeted treatment by leveraging the patient's immune system or adoptive transfer of active immune cells to selectively eliminate cancer cells. Despite notable clinical successes, conventional immunotherapies face significant challenges stemming from the poor infiltration of endogenous or adoptively transferred cytotoxic T cells in tumors, immunosuppressive tumor microenvironment and the immune evasion capability of cancer cells, leading to limited efficacy in many types of solid tumors. Overcoming these hurdles is essential to broaden the applicability of immunotherapies. Recent advances in nanotherapeutics have emerged as an innovative tool to overcome these challenges and enhance the therapeutic potential of tumor immunotherapy. The unique biochemical and biophysical properties of nanomaterials offer advantages in activation of immune cells in vitro for cell therapy, targeted delivery, and controlled release of immunomodulatory agents in vivo. Nanoparticles are excellent carriers for tumor associated antigens or neoantigen peptides for tumor vaccine, empowering activation of tumor specific T cell responses. By precisely delivering immunomodulatory agents to the tumor site, immunoactivating nanoparticles can promote tumor infiltration of endogenous T cells or adoptively transferred T cells into tumors, to overcoming delivery and biological barriers in the tumor microenvironment, augmenting the immune system's ability to recognize and eliminate cancer cells. This review provides an overview of the current advances in immunotherapeutic approaches utilizing nanotechnology. With a focus on discussions concerning strategies to enhance activity and efficacy of cytotoxic T cells and explore the intersection of engineering nanoparticles and immunomodulation aimed at bolstering T cell-mediated immune responses, we introduce various nanoparticle formulations designed to deliver therapeutic payloads, tumor antigens and immunomodulatory agents for T cell activation. Diverse mechanisms through which nanoparticle-based approaches influence T cell responses by improving antigen presentation, promoting immune cell trafficking, and reprogramming immunosuppressive tumor microenvironments to potentiate anti-tumor immunity are examined. Additionally, the synergistic potential of combining nanotherapeutics with existing immunotherapies, such as immune checkpoint inhibitors and adoptive T cell therapies is explored. In conclusion, this review highlights emerging research advances on activation of cytotoxic T cells using nanoparticle agents to support the promises and potential applications of nanoparticle-based immunomodulatory agents for cancer immunotherapy.
Collapse
Affiliation(s)
- Kory Wells
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tongrui Liu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lei Zhu
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Winship Cancer Institute, Clinic C, Room 4088, 1365 C Clifton Road, NE, Atlanta, GA 30322, USA.
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Yang H, Sun L, Chen R, Xiong Z, Yu W, Liu Z, Chen H. Biomimetic dendritic polymeric microspheres induce enhanced T cell activation and expansion for adoptive tumor immunotherapy. Biomaterials 2023; 296:122048. [PMID: 36842237 DOI: 10.1016/j.biomaterials.2023.122048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/23/2023]
Abstract
A variety of bioactive materials are currently developed to expand T cells ex vivo for adoptive T cell immunotherapy, also known as called artificial antigen-presenting cells (aAPCs). However, almost all the reported designs exhibit relatively smooth surface modified with T cell activating biomolecules, and therefore cannot well mimic the dendritic morphological characteristics of dendritic cells (DCs), the most important type of natural antigen-presenting cells (APCs) with high specific surface areas. Here, we propose a hydrophilic monomer-mediated surface morphology control strategy to synthesize biocompatible dendritic poly(N-isopropylacrylamide) (PNIPAM) microspheres for constructing aAPCs with surface morphology mimicking natural APCs (e.g., DCs). Interestingly, when maintaining the same ligands density, dendritic polymeric microspheres-based aAPCs (DPM beads) can more efficiently expand CD8+ T cells than that with smooth surfaces. Moreover, adoptive transfer of antigen-specific CD8+ T cells expanded by the DPM beads show significant antitumor effect of B16-OVA tumor bearing mice. Therefore, we provide a new concept for constructing biomimetic aAPCs with enhanced T cell expansion ability.
Collapse
Affiliation(s)
- He Yang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Lele Sun
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Rui Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Zijian Xiong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Wenzhuo Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China.
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
6
|
Zang H, Siddiqui M, Gummuluru S, Wong WW, Reinhard BM. Ganglioside-Functionalized Nanoparticles for Chimeric Antigen Receptor T-Cell Activation at the Immunological Synapse. ACS NANO 2022; 16:18408-18420. [PMID: 36282488 PMCID: PMC9815837 DOI: 10.1021/acsnano.2c06516] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has proven to be an effective strategy against hematological malignancies but persistence and activity against solid tumors must be further improved. One emerging strategy for enhancing efficacy is based on directing CAR T cells to antigen presenting cells (APCs). Activation of CAR T cells at the immunological synapse (IS) formed between APC and T cell is thought to promote strong, persistent antigen-specific T cell-mediated immune responses but requires integration of CAR ligands into the APC/T-cell interface. Here, we demonstrate that CAR ligand functionalized, lipid-coated, biodegradable polymer nanoparticles (NPs) that contain the ganglioside GM3 (GM3-NPs) bind to CD169 (Siglec-1)-expressing APCs and localize to the cell contact site between APCs and CAR T cells upon initiation of cell conjugates. The CD169+ APC/CAR T-cell interface is characterized by a strong optical colocalization of GM3-NPs and CARs, enrichment of F-actin, and recruitment of ZAP-70, indicative of integration of GM3-NPs into a functional IS. Ligands associated with GM3-NPs localized to the APC/T-cell contact site remain accessible to CARs and result in robust T-cell activation. Overall, this work identifies GM3-NPs as a potential antigen delivery platform for active targeting of CD169 expressing APCs and enhancement of CAR T-cell activation at the NP-containing IS.
Collapse
Affiliation(s)
- Han Zang
- Departments of Chemistry and The Photonics Center, Boston University, Boston, MA, 02215, United States
| | - Menna Siddiqui
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Suryaram Gummuluru
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, United States
| | - Wilson W. Wong
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, 02215, USA
| | - Björn M. Reinhard
- Departments of Chemistry and The Photonics Center, Boston University, Boston, MA, 02215, United States
| |
Collapse
|
7
|
Wauters A, Scheerstra JF, Vermeijlen IG, Hammink R, Schluck M, Woythe L, Wu H, Albertazzi L, Figdor CG, Tel J, Abdelmohsen LKEA, van Hest JCM. Artificial Antigen-Presenting Cell Topology Dictates T Cell Activation. ACS NANO 2022; 16:15072-15085. [PMID: 35969506 PMCID: PMC9527792 DOI: 10.1021/acsnano.2c06211] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/09/2022] [Indexed: 06/10/2023]
Abstract
Nanosized artificial antigen-presenting cells (aAPCs), synthetic immune cell mimics that aim to activate T cells ex or in vivo, offer an effective alternative to cellular immunotherapies. However, comprehensive studies that delineate the effect of nano-aAPC topology, including nanoparticle morphology and ligand density, are lacking. Here, we systematically studied the topological effects of polymersome-based aAPCs on T cell activation. We employed an aAPC library created from biodegradable poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-PDLLA) polymersomes with spherical or tubular shape and variable sizes, which were functionalized with αCD3 and αCD28 antibodies at controlled densities. Our results indicate that high ligand density leads to enhancement in T cell activation, which can be further augmented by employing polymersomes with larger size. At low ligand density, the effect of both polymersome shape and size was more pronounced, showing that large elongated polymersomes better activate T cells compared to their spherical or smaller counterparts. This study demonstrates the capacity of polymersomes as aAPCs and highlights the role of topology for their rational design.
Collapse
Affiliation(s)
- Annelies
C. Wauters
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jari F. Scheerstra
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Irma G. Vermeijlen
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Laura Woythe
- Department
of Biomedical Engineering, Institute of Complex Molecular Systems
(ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Hanglong Wu
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lorenzo Albertazzi
- Department
of Biomedical Engineering, Institute of Complex Molecular Systems
(ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08036, Spain
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, The Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, The Netherlands
| | - Jurjen Tel
- Department
of Biomedical Engineering, Institute of Complex Molecular Systems
(ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Laboratory
of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Loai K. E. A. Abdelmohsen
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Jan C. M. van Hest
- Bio-Organic
Chemistry, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
8
|
Sun L, Shen F, Xiong Z, Yang H, Dong Z, Xiang J, Gu Q, Ji Q, Fan C, Liu Z. DNA Engineered Lymphocyte-Based Homologous Targeting Artificial Antigen-Presenting Cells for Personalized Cancer Immunotherapy. J Am Chem Soc 2022; 144:7634-7645. [PMID: 35438987 DOI: 10.1021/jacs.1c09316] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Artificial antigen-presenting cells (aAPCs) constructed by integrating T cell activation ligands on biocompatible materials hold great potential in tumor immunotherapy. However, it remains challenging to develop aAPCs, which could mimic the characteristics of natural APCs, thereby realizing antigen-specific T cells activation in vivo. Here, we report the first effort to construct natural lymphocyte-based homologous targeting aAPCs (LC-aAPCs) with lipid-DNA-mediated noninvasive live cell surface engineering. Through a predesigned bottom-up self-assembly path, we achieved natural-APC-mimicking distribution of T cell activation ligands on LC-aAPCs, which would enable the optimized T cell activation. Moreover, the lipid-DNA-mediated self-assembly occurring on lipid bilayers would not affect the functions of homing receptors expressed on lymphocyte. Therefore, such LC-aAPCs could actively migrate to peripheral lymphatic organs and then effectively activate antigen-specific T cells. Combined with an immune checkpoint inhibitor, such LC-aAPCs could effectively inhibit the growth of different tumor models. Thus, our work provides a new design of aAPCs for in vivo applications in tumor immunotherapy, and the lipid-DNA-mediated noninvasive live cell surface engineering would be a powerful tool for designing cell-based therapeutics.
Collapse
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zijian Xiong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - He Yang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jian Xiang
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qingyang Gu
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qunsheng Ji
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Chunhai Fan
- Natl Ctr Translat Med, Frontiers Sci Ctr Transformat Mol, Sch Chem & Chem Engn, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
9
|
Ukrainskaya V, Rubtsov Y, Pershin D, Podoplelova N, Terekhov S, Yaroshevich I, Sokolova A, Bagrov D, Kulakovskaya E, Shipunova V, Deyev S, Ziganshin R, Chernov A, Telegin G, Maksimov E, Markov O, Oshchepkova A, Zenkova M, Xie J, Zhang H, Gabibov A, Maschan M, Stepanov A, Lerner R. Antigen-Specific Stimulation and Expansion of CAR-T Cells Using Membrane Vesicles as Target Cell Surrogates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102643. [PMID: 34605165 DOI: 10.1002/smll.202102643] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/29/2021] [Indexed: 06/13/2023]
Abstract
Development of CAR-T therapy led to immediate success in the treatment of B cell leukemia. Manufacturing of therapy-competent functional CAR-T cells needs robust protocols for ex vivo/in vitro expansion of modified T-cells. This step is challenging, especially if non-viral low-efficiency delivery protocols are used to generate CAR-T cells. Modern protocols for CAR-T cell expansion are imperfect since non-specific stimulation results in rapid outgrowth of CAR-negative T cells, and removal of feeder cells from mixed cultures necessitates additional purification steps. To develop a specific and improved protocol for CAR-T cell expansion, cell-derived membrane vesicles are taken advantage of, and the simple structural demands of the CAR-antigen interaction. This novel approach is to make antigenic microcytospheres from common cell lines stably expressing surface-bound CAR antigens, and then use them for stimulation and expansion of CAR-T cells. The data presented in this article clearly demonstrate that this protocol produced antigen-specific vesicles with the capacity to induce stronger stimulation, proliferation, and functional activity of CAR-T cells than is possible with existing protocols. It is predicted that this new methodology will significantly advance the ability to obtain improved populations of functional CAR-T cells for therapy.
Collapse
Affiliation(s)
- Valeria Ukrainskaya
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Yuri Rubtsov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Dmitry Pershin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Nadezhda Podoplelova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Stanislav Terekhov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Igor Yaroshevich
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - Anstasiia Sokolova
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya, 1a, Moscow, 119435, Russia
| | - Dmitry Bagrov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - Elena Kulakovskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Victoria Shipunova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Sergey Deyev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Rustam Ziganshin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Aleksandr Chernov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Georgii Telegin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Eugene Maksimov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - Oleg Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave. 8, Novosibirsk, 630090, Russia
| | - Anastasiya Oshchepkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave. 8, Novosibirsk, 630090, Russia
| | - Marina Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrentiev Ave. 8, Novosibirsk, 630090, Russia
| | - Jia Xie
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road MB-10, La Jolla, CA, 92037, USA
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Alexander Gabibov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
| | - Michael Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
| | - Alexey Stepanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117997, Russia
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road MB-10, La Jolla, CA, 92037, USA
| | - Richard Lerner
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road MB-10, La Jolla, CA, 92037, USA
| |
Collapse
|
10
|
Peptide-HLA-based immunotherapeutics platforms for direct modulation of antigen-specific T cells. Sci Rep 2021; 11:19220. [PMID: 34584159 PMCID: PMC8479091 DOI: 10.1038/s41598-021-98716-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/06/2021] [Indexed: 11/20/2022] Open
Abstract
Targeted pharmacologic activation of antigen-specific (AgS) T cells may bypass limitations inherent in current T cell-based cancer therapies. We describe two immunotherapeutics platforms for selective delivery of costimulatory ligands and peptide-HLA (pHLA) to AgS T cells. We engineered and deployed on these platforms an affinity-attenuated variant of interleukin-2, which selectively expands oligoclonal and polyfunctional AgS T cells in vitro and synergizes with CD80 signals for superior proliferation versus peptide stimulation.
Collapse
|
11
|
Bacterial Outer Membrane Vesicles as a Versatile Tool in Vaccine Research and the Fight against Antimicrobial Resistance. mBio 2021; 12:e0170721. [PMID: 34372691 PMCID: PMC8406158 DOI: 10.1128/mbio.01707-21] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gram-negative bacteria include a number of pathogens that cause disease in humans and animals. Although antibiotics are still effective in treating a considerable range of infections caused by Gram-negative bacteria, the alarming increase of antimicrobial resistance (AMR) induced by excessive use of antibiotics has raised global concerns. Therefore, alternative strategies must be developed to prevent and treat bacterial infections and prevent the advent of a postantibiotic era. Vaccines, one of the greatest achievements in the history of medical science, hold extraordinary potential to prevent bacterial infections and thereby reduce the need for antibiotics. Novel bacterial vaccines are urgently needed, however, and outer membrane vesicles (OMVs), naturally produced by Gram-negative bacteria, represent a promising and versatile tool that can be employed as adjuvants, antigens, and delivery platforms in the development of vaccines against Gram-negative bacteria. Here, we provide an overview of the many roles OMVs can play in vaccine development and the mechanisms behind these applications. Methods to improve OMV yields and a comparison of different strategies for OMV isolation aiming at cost-effective production of OMV-based vaccines are also reviewed.
Collapse
|
12
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Zhang X, Luo M, Dastagir SR, Nixon M, Khamhoung A, Schmidt A, Lee A, Subbiah N, McLaughlin DC, Moore CL, Gribble M, Bayhi N, Amin V, Pepi R, Pawar S, Lyford TJ, Soman V, Mellen J, Carpenter CL, Turka LA, Wickham TJ, Chen TF. Engineered red blood cells as an off-the-shelf allogeneic anti-tumor therapeutic. Nat Commun 2021; 12:2637. [PMID: 33976146 PMCID: PMC8113241 DOI: 10.1038/s41467-021-22898-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/31/2021] [Indexed: 02/03/2023] Open
Abstract
Checkpoint inhibitors and T-cell therapies have highlighted the critical role of T cells in anti-cancer immunity. However, limitations associated with these treatments drive the need for alternative approaches. Here, we engineer red blood cells into artificial antigen-presenting cells (aAPCs) presenting a peptide bound to the major histocompatibility complex I, the costimulatory ligand 4-1BBL, and interleukin (IL)-12. This leads to robust, antigen-specific T-cell expansion, memory formation, additional immune activation, tumor control, and antigen spreading in tumor models in vivo. The presence of 4-1BBL and IL-12 induces minimal toxicities due to restriction to the vasculature and spleen. The allogeneic aAPC, RTX-321, comprised of human leukocyte antigen-A*02:01 presenting the human papilloma virus (HPV) peptide HPV16 E711-19, 4-1BBL, and IL-12 on the surface, activates HPV-specific T cells and promotes effector function in vitro. Thus, RTX-321 is a potential 'off-the-shelf' in vivo cellular immunotherapy for treating HPV + cancers, including cervical and head/neck cancers.
Collapse
Affiliation(s)
- Xuqing Zhang
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Mengyao Luo
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Shamael R. Dastagir
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Mellissa Nixon
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Annie Khamhoung
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Andrea Schmidt
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Albert Lee
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Naren Subbiah
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | | | | | - Mary Gribble
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Nicholas Bayhi
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Viral Amin
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Ryan Pepi
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Sneha Pawar
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Timothy J. Lyford
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Vikram Soman
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Jennifer Mellen
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | | | - Laurence A. Turka
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Thomas J. Wickham
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| | - Tiffany F. Chen
- grid.507501.60000 0004 6022 070XRubius Therapeutics, Inc., Cambridge, MA USA
| |
Collapse
|
14
|
Campbell EA, Ramirez K, Holegadde M, Yeshlur N, Khaja A, Sulchek TA. Tuning Antibody Presentation to Enhance T-Cell Activation for Downstream Cytotoxicity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:4783-4792. [PMID: 33848167 DOI: 10.1021/acs.langmuir.0c03203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cytotoxic effector cells are an integral component of the immune response against pathogens and diseases such as cancer and thus of great interest to researchers who wish to enhance the native immune response. Although researchers routinely use particles to stimulate cytotoxic T cells, few studies have comprehensively investigated: (1) beyond initial activation responses (i.e., proliferation and CD25/CD69 expression) to downstream cancer-killing effects and (2) how to drive cytotoxic T-cell responses by adjusting biomolecular and physical properties of particles. In this study, we designed particles displaying an anti-CD3 antibody to activate cytotoxic T cells and study their downstream cytotoxic effects. We evaluated the effect of antibody immobilization, particle size, molecular surface density of an anti-CD3 antibody, and the inclusion of an anti-CD28 antibody on cytolytic granule release by T cells. We found that immobilizing the anti-CD3 antibody onto smaller nanoparticles elicited increased T-cell activation products for an equivalent delivery of the anti-CD3 antibody. We further established that the mechanism behind increased cancer cell death was associated with the proximity of T cells to cancer cells. Functionalizing particles additionally with the anti-CD28 antibody at an optimized antibody density caused increased T-cell proliferation and T-cell binding but we observed no effective increase in cytotoxicity. Meaningfully, our results are discussed within the context of commercially available and widely used anti-CD3/28 Dynabeads. These results showed that T-cell activation and cytotoxicity can be optimized with a molecular presentation on smaller particles and thus, offer exciting new possibilities to engineer T-cell activation responses for effective outcomes.
Collapse
Affiliation(s)
- Elizabeth A Campbell
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Katily Ramirez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Meghana Holegadde
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Nayana Yeshlur
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Akram Khaja
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Todd A Sulchek
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30313, United States
| |
Collapse
|
15
|
Hammink R, Weiden J, Voerman D, Popelier C, Eggermont LJ, Schluck M, Figdor CG, Verdoes M. Semiflexible Immunobrushes Induce Enhanced T Cell Activation and Expansion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16007-16018. [PMID: 33797875 PMCID: PMC8045021 DOI: 10.1021/acsami.0c21994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A variety of bioactive materials developed to expand T cells for adoptive transfer into cancer patients are currently evaluated in the clinic. In most cases, T cell activating biomolecules are attached to rigid surfaces or matrices and form a static interface between materials and the signaling receptors on the T cells. We hypothesized that a T cell activating polymer brush interface might better mimic the cell surface of a natural antigen-presenting cell, facilitating receptor movement and concomitant advantageous mechanical forces to provide enhanced T cell activating capacities. Here, as a proof of concept, we synthesized semiflexible polyisocyanopeptide (PIC) polymer-based immunobrushes equipped with T cell activating agonistic anti-CD3 (αCD3) and αCD28 antibodies placed on magnetic microbeads. We demonstrated enhanced efficiency of ex vivo expansion of activated primary human T cells even at very low numbers of stimulating antibodies compared to rigid beads. Importantly, the immunobrush architecture appeared crucial for this improved T cell activating capacity. Immunobrushes outperform current benchmarks by producing higher numbers of T cells exhibiting a combination of beneficial phenotypic characteristics, such as reduced exhaustion marker expression, high cytokine production, and robust expression of cytotoxic hallmarks. This study indicates that semiflexible immunobrushes have great potential in making T cell-based immunotherapies more effective.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jorieke Weiden
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Dion Voerman
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carlijn Popelier
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Martijn Verdoes
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| |
Collapse
|
16
|
Raza F, Zafar H, Zhang S, Kamal Z, Su J, Yuan W, Mingfeng Q. Recent Advances in Cell Membrane-Derived Biomimetic Nanotechnology for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2002081. [PMID: 33586322 DOI: 10.1002/adhm.202002081] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/13/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy will significantly impact the standard of care in cancer treatment. Recent advances in nanotechnology can improve the efficacy of cancer immunotherapy. However, concerns regarding efficiency of cancer nanomedicine, complex tumor microenvironment, patient heterogeneity, and systemic immunotoxicity drive interest in more novel approaches to be developed. For this purpose, biomimetic nanoparticles are developed to make innovative changes in the delivery and biodistribution of immunotherapeutics. Biomimetic nanoparticles have several advantages that can advance the clinical efficacy of cancer immunotherapy. Thus there is a greater push toward the utilization of biomimetic nanotechnology for developing effective cancer immunotherapeutics that demonstrate increased specificity and potency. The recent works and state-of-the-art strategies for anti-tumor immunotherapeutics are highlighted here, and particular emphasis has been given to the applications of cell-derived biomimetic nanotechnology for cancer immunotherapy.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Hajra Zafar
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Shulei Zhang
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Zul Kamal
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
- Department of Pharmacy Shaheed Benazir Bhutto University Sheringal Dir (Upper) Khyber Pakhtunkhwa 18000 Pakistan
| | - Jing Su
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Wei‐En Yuan
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| | - Qiu Mingfeng
- School of Pharmacy Shanghai Jiao Tong University Shanghai 200240 P. R. China
| |
Collapse
|
17
|
Sun L, Shen F, Xu J, Han X, Fan C, Liu Z. DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202003367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Shanghai Jiao Tong University Shanghai 201240 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| |
Collapse
|
18
|
DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020; 59:14842-14853. [DOI: 10.1002/anie.202003367] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/25/2020] [Indexed: 12/21/2022]
|
19
|
Nanoparticle mediated cancer immunotherapy. Semin Cancer Biol 2020; 69:307-324. [PMID: 32259643 DOI: 10.1016/j.semcancer.2020.03.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
The versatility and nanoscale size have helped nanoparticles (NPs) improve the efficacy of conventional cancer immunotherapy and opened up exciting approaches to combat cancer. This review first outlines the tumor immune evasion and the defensive tumor microenvironment (TME) that hinders the activity of host immune system against tumor. Then, a detailed description on how the NP based strategies have helped improve the efficacy of conventional cancer vaccines and overcome the obstacles led by TME. Sustained and controlled drug delivery, enhanced cross presentation by immune cells, co-encapsulation of adjuvants, inhibition of immune checkpoints and intrinsic adjuvant like properties have aided NPs to improve the therapeutic efficacy of cancer vaccines. Also, NPs have been efficient modulators of TME. In this context, NPs facilitate better penetration of the chemotherapeutic drug by dissolution of the inhibitory meshwork formed by tumor associated cells, blood vessels, soluble mediators and extra cellular matrix in TME. NPs achieve this by suppression, modulation, or reprogramming of the immune cells and other mediators localised in TME. This review further summarizes the applications of NPs used to enhance the efficacy of cancer vaccines and modulate the TME to improve cancer immunotherapy. Finally, the hurdles faced in commercialization and translation to clinic have been discussed and intriguingly, NPs owe great potential to emerge as clinical formulations for cancer immunotherapy in near future.
Collapse
|
20
|
Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: opportunities and challenges. NANOSCALE 2020; 12:5746-5763. [PMID: 32124894 DOI: 10.1039/c9nr08958f] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines harness the inherent properties of the immune system to prevent diseases or treat existing ones. Continuous efforts have been devoted to both gaining a mechanistic understanding of how the immune system operates and designing vaccines with high efficacies and effectiveness. Advancements in nanotechnology in recent years have generated unique opportunities to meet the daunting challenges associated with immunology and vaccine development. Firstly, nanoparticle formulated systems provide ideal model systems for studying the operation of the immune system, making it possible to systematically identify key factors and understand their roles in specific immune responses. Also, the versatile compositions/architectures of nanoparticle systems enable new strategies/novel platforms for developing vaccines with high efficacies and effectiveness. In this review, we discuss the advantages of nanoparticles and the challenges faced during vaccine development, through the framework of the immunological mechanisms of vaccination, with the aim of bridging the gap between immunology and materials science, which are both involved in vaccine design. The knowledge obtained provides general guidelines for future vaccine development.
Collapse
Affiliation(s)
- Pingsai Lung
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | |
Collapse
|
21
|
Majedi FS, Hasani-Sadrabadi MM, Thauland TJ, Li S, Bouchard LS, Butte MJ. Augmentation of T-Cell Activation by Oscillatory Forces and Engineered Antigen-Presenting Cells. NANO LETTERS 2019; 19:6945-6954. [PMID: 31478664 PMCID: PMC6786928 DOI: 10.1021/acs.nanolett.9b02252] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Activation of T cells by antigen presenting cells (APCs) initiates their proliferation, cytokine production, and killing of infected or cancerous cells. We and others have shown that T-cell receptors require mechanical forces for triggering, and these forces arise during the interaction of T cells with APCs. Efficient activation of T cells in vitro is necessary for clinical applications. In this paper, we studied the impact of combining mechanical, oscillatory movements provided by an orbital shaker with soft, biocompatible, artificial APCs (aAPCs) of various sizes and amounts of antigen. We showed that these aAPCs allow for testing the strength of signal delivered to T cells, and enabled us to confirm that that absolute amounts of antigen engaged by the T cell are more important for activation than the density of antigen. We also found that when our aAPCs interact with T cells in the context of an oscillatory mechanoenvironment, they roughly double antigenic signal strength, compared to conventional, static culture. Combining these effects, our aAPCs significantly outperformed the commonly used Dynabeads. We finally demonstrated that tuning the signal strength down to a submaximal "sweet spot" allows for robust expansion of induced regulatory T cells. In conclusion, augmenting engineered aAPCs with mechanical forces offers a novel approach for tuning of T-cell activation and differentiation.
Collapse
Affiliation(s)
- Fatemeh S. Majedi
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095, United States
| | | | - Timothy J. Thauland
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Louis-S. Bouchard
- Department of Bioengineering, University of California Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Manish J. Butte
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, University of California Los Angeles, Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California 90095, United States
- Corresponding Author: Tel.: 310-825-6482. Fax: 310-825-9832. . Address: Department of Pediatrics, UCLA, 10833 Le Conte Ave., MDCC Building Room 12-430, Los Angeles, CA 90095, USA
| |
Collapse
|
22
|
Zhang L, Song S, Jin X, Wan X, Shahzad KA, Pei W, Zhao C, Shen C. An Artificial Antigen-Presenting Cell Delivering 11 Immune Molecules Expands Tumor Antigen–Specific CTLs in Ex Vivo and In Vivo Murine Melanoma Models. Cancer Immunol Res 2019; 7:1188-1201. [DOI: 10.1158/2326-6066.cir-18-0881] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/19/2019] [Accepted: 05/17/2019] [Indexed: 11/16/2022]
|
23
|
Song S, Jin X, Zhang L, Zhao C, Ding Y, Ang Q, Khaidav O, Shen C. PEGylated and CD47-conjugated nanoellipsoidal artificial antigen-presenting cells minimize phagocytosis and augment anti-tumor T-cell responses. Int J Nanomedicine 2019; 14:2465-2483. [PMID: 31040669 PMCID: PMC6459144 DOI: 10.2147/ijn.s195828] [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] [Indexed: 11/23/2022] Open
Abstract
Purpose Antigen-presenting cells (APCs) are powerful tools to expand antigen-specific T cells ex vivo and in vivo for tumor immunotherapy, but suffer from time-consuming generation and biosafety concerns raised by live cells. Alternatively, the cell-free artificial antigen-presenting cells (aAPCs) have been rapidly developed. Nanoscale aAPCs are recently proposed owing to their superior biodistribution and reduced embolism than conventional cell-sized aAPCs, but pose the challenges: easier cellular uptake and smaller contact surface area with T cells than the cell-sized counterparts. This study aimed to fabricate a new “stealth” nano-aAPCs with microscale contact surface area to minimize cellular uptake and activate antigen-specific T cells by combination uses of ellipsoidal stretch, PEGylation, and self-marker CD47-Fc conjugation. Methods The spherical polylactic-co-glycolic acid nanoparticles were fabricated using a double-emulsion method, and then stretched twofold using film-stretching procedure followed by PEGylation and co-coupling with CD47-Fc, H-2Kb/TRP2180-188-Ig dimers, and anti-CD28. The resulting PEGylated and CD47-conjugated nanoellipsoidal aAPCs (EaAPCPEG/CD47) were co-cultured with macrophages or spleen lymphocytes and also infused into melanoma-bearing mice. The in vitro and in vivo effects were evaluated and compared with the nanospherical aAPCs (SaAPC), nanoellipsoidal aAPCs (EaAPC), or PEGylated nanoellipsoidal aAPC (EaAPCPEG). Results EaAPCPEG/CD47 markedly reduced cellular uptake in vitro and in vivo, as compared with EaAPCPEG, EaAPC, SaAPC, and Blank-NPs and expanded naïve TRP2180-188-specific CD8+ T cells in the co-cultures with spleen lymphocytes. After three infusions, the EaAPCPEG/CD47 showed much stronger effects on facilitating TRP2180-188-specific CD8+ T-cell proliferation, local infiltration, and tumor necrosis in the melanoma-bearing mice and on inhibiting tumor growth than the control aAPCs. Conclusion The superimposed or synergistic effects of ellipsoidal stretch, PEGylation, and CD47-Fc conjugation minimized cellular uptake of nano-aAPCs and enhanced their functionality to expand antigen-specific T cells and inhibit tumor growth, thus suggesting a more valuable strategy to design “stealth” nanoscale aAPCs suitable for tumor active immunotherapy.
Collapse
Affiliation(s)
- Shilong Song
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Xiaoxiao Jin
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Chen Zhao
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Yan Ding
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Qianqian Ang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Odontuya Khaidav
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China,
| |
Collapse
|
24
|
Olden BR, Perez CR, Wilson AL, Cardle II, Lin YS, Kaehr B, Gustafson JA, Jensen MC, Pun SH. Cell-Templated Silica Microparticles with Supported Lipid Bilayers as Artificial Antigen-Presenting Cells for T Cell Activation. Adv Healthc Mater 2019; 8:e1801188. [PMID: 30549244 PMCID: PMC6394850 DOI: 10.1002/adhm.201801188] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/28/2018] [Indexed: 01/18/2023]
Abstract
Biomaterial properties that modulate T cell activation, growth, and differentiation are of significant interest in the field of cellular immunotherapy manufacturing. In this work, a new platform technology that allows for the modulation of various activation particle design parameters important for polyclonal T cell activation is presented. Artificial antigen presenting cells (aAPCs) are successfully created using supported lipid bilayers on various cell-templated silica microparticles with defined membrane fluidity and stimulating antibody density. This panel of aAPCs is used to probe the importance of activation particle shape, size, membrane fluidity, and stimulation antibody density on T cell outgrowth and differentiation. All aAPC formulations are able to stimulate T cell growth, and preferentially promote CD8+ T cell growth over CD4+ T cell growth when compared to commercially available pendant antibody-conjugated particles. T cells cultured with HeLa- and red blood cell-templated aAPCs have a less-differentiated and less-exhausted phenotype than those cultured with spherical aAPCs with matched membrane coatings when cultured for 14 days. These results support continued exploration of silica-supported lipid bilayers as an aAPC platform.
Collapse
Affiliation(s)
- Brynn R. Olden
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Caleb R. Perez
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Ashley L. Wilson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Ian I. Cardle
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Yu-Shen Lin
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| | - Bryan Kaehr
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, NM 87185, USA
| | - Joshua A. Gustafson
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Michael C. Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Suzie H. Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195, USA,
| |
Collapse
|
25
|
Schappert A, Schneck JP, Suarez L, Oelke M, Schütz C. Soluble MHC class I complexes for targeted immunotherapy. Life Sci 2018; 209:255-258. [PMID: 30102903 DOI: 10.1016/j.lfs.2018.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/27/2022]
Abstract
Major histocompatibility complexes (MHC) have been used for more than two decades in clinical and pre-clinical approaches of tumor immunotherapy. They have been proven efficient for detecting anti-tumor-specific T cells when utilized as soluble multimers, immobilized on cells or artificial structures such as artificial antigen-presenting cells (aAPC) and have been shown to generate effective anti-tumor responses. In this review we summarize the use of soluble MHC class I complexes in tumor vaccination studies, highlighting the different strategies and their contradicting results. In summary, we believe that soluble MHC class I molecules represent an exciting tool with great potential to impact the understanding and development of immunotherapeutic approaches on many levels from monitoring to treatment.
Collapse
Affiliation(s)
- Anna Schappert
- Paul-Ehrlich-Institute, Division of Immunology, Langen, Germany; Medical Clinic 1, University Hospital/Goethe University of Frankfurt am Main, Germany.
| | - Jonathan P Schneck
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
26
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Smith MR, Tolbert SV, Wen F. Protein-Scaffold Directed Nanoscale Assembly of T Cell Ligands: Artificial Antigen Presentation with Defined Valency, Density, and Ratio. ACS Synth Biol 2018; 7:1629-1639. [PMID: 29733631 DOI: 10.1021/acssynbio.8b00119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tuning antigen presentation to T cells is a critical step in investigating key aspects of T cell activation. However, existing technologies have a limited ability to control the spatial and stoichiometric organization of T cell ligands on 3D surfaces. Here, we developed an artificial antigen presentation platform based on protein scaffold-directed assembly that allows fine control over the spatial and stoichiometric organization of T cell ligands on a 3D yeast cell surface. Using this system, we observed that the T cell activation threshold on a 3D surface is independent of peptide-major histocompatibility complex (pMHC) valency but instead is determined by the overall pMHC surface density. When intercellular adhesion molecule 1 (ICAM-1) was coassembled with pMHC, it enhanced antigen recognition sensitivity by 6-fold. Further, T cells responded with different magnitudes to varying ratios of pMHC and ICAM-1 and exhibited a maximum response at a ratio of 15% pMHC and 85% ICAM-1, introducing an additional parameter for tuning T cell activation. This protein scaffold-directed assembly technology is readily transferrable to acellular surfaces for translational research as well as large-scale T-cell manufacturing.
Collapse
Affiliation(s)
- Mason R. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Stephanie V. Tolbert
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
28
|
de la Zerda A, Kratochvil MJ, Suhar NA, Heilshorn SC. Review: Bioengineering strategies to probe T cell mechanobiology. APL Bioeng 2018; 2:021501. [PMID: 31069295 PMCID: PMC6324202 DOI: 10.1063/1.5006599] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/29/2018] [Indexed: 01/08/2023] Open
Abstract
T cells play a major role in adaptive immune response, and T cell dysfunction can lead to the progression of several diseases that are often associated with changes in the mechanical properties of tissues. However, the concept that mechanical forces play a vital role in T cell activation and signaling is relatively new. The endogenous T cell microenvironment is highly complex and dynamic, involving multiple, simultaneous cell-cell and cell-matrix interactions. This native complexity has made it a challenge to isolate the effects of mechanical stimuli on T cell activation. In response, researchers have begun developing engineered platforms that recapitulate key aspects of the native microenvironment to dissect these complex interactions in order to gain a better understanding of T cell mechanotransduction. In this review, we first describe some of the unique characteristics of T cells and the mounting research that has shown they are mechanosensitive. We then detail the specific bioengineering strategies that have been used to date to measure and perturb the mechanical forces at play during T cell activation. In addition, we look at engineering strategies that have been used successfully in mechanotransduction studies for other cell types and describe adaptations that may make them suitable for use with T cells. These engineering strategies can be classified as 2D, so-called 2.5D, or 3D culture systems. In the future, findings from this emerging field will lead to an optimization of culture environments for T cell expansion and the development of new T cell immunotherapies for cancer and other immune diseases.
Collapse
Affiliation(s)
- Adi de la Zerda
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | | | - Nicholas A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
29
|
Kosmides AK, Necochea K, Hickey JW, Schneck JP. Separating T Cell Targeting Components onto Magnetically Clustered Nanoparticles Boosts Activation. NANO LETTERS 2018; 18:1916-1924. [PMID: 29488768 PMCID: PMC6707078 DOI: 10.1021/acs.nanolett.7b05284] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
T cell activation requires the coordination of a variety of signaling molecules including T cell receptor-specific signals and costimulatory signals. Altering the composition and distribution of costimulatory molecules during stimulation greatly affects T cell functionality for applications such as adoptive cell therapy (ACT), but the large diversity in these molecules complicates these studies. Here, we develop and validate a reductionist T cell activation platform that enables streamlined customization of stimulatory conditions. This platform is useful for the optimization of ACT protocols as well as the more general study of immune T cell activation. Rather than decorating particles with both signal 1 antigen and signal 2 costimulus, we use distinct, monospecific, paramagnetic nanoparticles, which are then clustered on the cell surface by a magnetic field. This allows for rapid synthesis and characterization of a small number of single-signal nanoparticles which can be systematically combined to explore and optimize T cell activation. By increasing cognate T cell enrichment and incorporating additional costimulatory molecules using this platform, we find significantly higher frequencies and numbers of cognate T cells stimulated from an endogenous population. The magnetic field-induced association of separate particles thus provides a tool for optimizing T cell activation for adoptive immunotherapy and other immunological studies.
Collapse
Affiliation(s)
- Alyssa K. Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Kevin Necochea
- Department of Materials Science and Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - John W. Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Jonathan P. Schneck
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Corresponding Author:
| |
Collapse
|
30
|
Abstract
Over the last century, there has been a dramatic change in the nature of therapeutic, biologically active molecules available to treat disease. Therapies have evolved from extracted natural products towards rationally designed biomolecules, including small molecules, engineered proteins and nucleic acids. The use of potent drugs which target specific organs, cells or biochemical pathways, necessitates new tools which can enable controlled delivery and dosing of these therapeutics to their biological targets. Here, we review the miniaturisation of drug delivery systems from the macro to nano-scale, focussing on controlled dosing and controlled targeting as two key parameters in drug delivery device design. We describe how the miniaturisation of these devices enables the move from repeated, systemic dosing, to on-demand, targeted delivery of therapeutic drugs and highlight areas of focus for the future.
Collapse
Affiliation(s)
- Derfogail Delcassian
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,b Department of Anaesthesiology , Boston Children's Hospital, Harvard Medical School , Boston , MA , USA.,c Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Asha K Patel
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,d Division of Cancer and Stem Cells, School of Medicine, and Division of Advanced Materials and Healthcare Technologies, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Abel B Cortinas
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Robert Langer
- a David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , Cambridge , MA , USA.,e Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA.,f Institute for Medical Engineering and Science , Massachusetts Institute of Technology , Cambridge , MA , USA.,g Media Lab , Massachusetts Institute of Technology , Cambridge , MA , USA
| |
Collapse
|
31
|
Hickey JW, Vicente FP, Howard GP, Mao HQ, Schneck JP. Biologically Inspired Design of Nanoparticle Artificial Antigen-Presenting Cells for Immunomodulation. NANO LETTERS 2017; 17:7045-7054. [PMID: 28994285 PMCID: PMC6709596 DOI: 10.1021/acs.nanolett.7b03734] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Particles engineered to engage and interact with cell surface ligands and to modulate cells can be harnessed to explore basic biological questions as well as to devise cellular therapies. Biology has inspired the design of these particles, such as artificial antigen-presenting cells (aAPCs) for use in immunotherapy. While much has been learned about mimicking antigen presenting cell biology, as we decrease the size of aAPCs to the nanometer scale, we need to extend biomimetic design to include considerations of T cell biology-including T-cell receptor (TCR) organization. Here we describe the first quantitative analysis of particle size effect on aAPCs with both Signals 1 and 2 based on T cell biology. We show that aAPCs, larger than 300 nm, activate T cells more efficiently than smaller aAPCs, 50 nm. The 50 nm aAPCs require saturating doses or require artificial magnetic clustering to activate T cells. Increasing ligand density alone on the 50 nm aAPCs did not increase their ability to stimulate CD8+ T cells, confirming the size-dependent phenomenon. These data support the need for multireceptor ligation and activation of T-cell receptor (TCR) nanoclusters of similar sizes to 300 nm aAPCs. Quantitative analysis and modeling of a nanoparticle system provides insight into engineering constraints of aAPCs for T cell immunotherapy applications and offers a case study for other cell-modulating particles.
Collapse
Affiliation(s)
- John W. Hickey
- Department of Biomedical Engineering, School of Medicine
- Institute for Cell Engineering, School of Medicine
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
| | | | - Gregory P. Howard
- Department of Biomedical Engineering, School of Medicine
- Institute for Nanobiotechnology
| | - Hai-Quan Mao
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
- Department of Materials Science and Engineering, Whiting School of Engineering
| | - Jonathan P. Schneck
- Institute for Cell Engineering, School of Medicine
- Department of Pathology, School of Medicine
- Institute for Nanobiotechnology
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Corresponding Author: . Phone: 410-614-4589
| |
Collapse
|
32
|
Sun X, Han X, Xu L, Gao M, Xu J, Yang R, Liu Z. Surface-Engineering of Red Blood Cells as Artificial Antigen Presenting Cells Promising for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701864. [PMID: 28861943 DOI: 10.1002/smll.201701864] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Indexed: 06/07/2023]
Abstract
The development of artificial antigen presenting cells (aAPCs) to mimic the functions of APCs such as dendritic cells (DCs) to stimulate T cells and induce antitumor immune responses has attracted substantial interests in cancer immunotherapy. In this work, a unique red blood cell (RBC)-based aAPC system is designed by engineering antigen peptide-loaded major histocompatibility complex-I and CD28 activation antibody on RBC surface, which are further tethered with interleukin-2 (IL2) as a proliferation and differentiation signal. Such RBC-based aAPC-IL2 (R-aAPC-IL2) can not only provide a flexible cell surface with appropriate biophysical parameters, but also mimic the cytokine paracrine delivery. Similar to the functions of matured DCs, the R-aAPC-IL2 cells can facilitate the proliferation of antigen-specific CD8+ T cells and increase the secretion of inflammatory cytokines. As a proof-of-concept, we treated splenocytes from C57 mice with R-aAPC-IL2 and discovered those splenocytes induced significant cancer-cell-specific lysis, implying that the R-aAPC-IL2 were able to re-educate T cells and induce adoptive immune response. This work thus presents a novel RBC-based aAPC system which can mimic the functions of antigen presenting DCs to activate T cells, promising for applications in adoptive T cell transfer or even in direct activation of circulating T cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ligeng Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Min Gao
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Rong Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
33
|
Zhang L, Wang L, Shahzad KA, Xu T, Wan X, Pei W, Shen C. Paracrine release of IL-2 and anti-CTLA-4 enhances the ability of artificial polymer antigen-presenting cells to expand antigen-specific T cells and inhibit tumor growth in a mouse model. Cancer Immunol Immunother 2017; 66:1229-1241. [PMID: 28501941 PMCID: PMC11028408 DOI: 10.1007/s00262-017-2016-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 05/06/2017] [Indexed: 12/22/2022]
Abstract
Accumulating evidence indicates that bead-based artificial antigen-presenting cells (aAPCs) are a powerful tool to induce antigen-specific T cell responses in vitro and in vivo. To date, most conventional aAPCs have been generated by coupling an antigen signal (signal 1) and one or two costimulatory signals, such as anti-CD28 with anti-LFA1 or anti-4-1BB (signal 2), onto the surfaces of cell-sized or nanoscale magnetic beads or polyester latex beads. The development of a biodegradable scaffold and the combined use of multiple costimulatory signals as well as third signals for putative clinical applications is the next step in the development of this technology. Here, a novel biodegradable aAPC platform for active immunotherapy was developed by co-encapsulating IL-2 and anti-CTLA-4 inside cell-sized polylactic-co-glycolic acid microparticles (PLGA-MPs) while co-coupling an H-2Kb/TRP2-Ig dimer and anti-CD28 onto the surface. Cytokines (activating signal) and antibodies (anti-inhibition signal) were efficiently co-encapsulated in PLGA-MP-based aAPCs and co-released without interfering with each other. The targeted, sustained co-release of IL-2 and anti-CTLA-4 achieved markedly enhanced, synergistic effects in activating and expanding tumor antigen-specific T cells both in vitro and in vivo, as well as in inhibiting tumor growth in a mouse melanoma model, as compared with conventional two-signal aAPCs and IL-2 or anti-CTLA-4 single-released aAPCs. These data revealed the feasibility and importance of the paracrine release of multiple costimulatory molecules and cytokines from biodegradable aAPCs and thus provide a proof of principle for the future use of polymeric aAPCs for active immunotherapy of tumors and infectious diseases.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Limin Wang
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Tao Xu
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Xin Wan
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Weiya Pei
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University, 87 Dingjiaqiao Rd, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
34
|
Wang C, Sun W, Ye Y, Bomba HN, Gu Z. Bioengineering of Artificial Antigen Presenting Cells and Lymphoid Organs. Theranostics 2017; 7:3504-3516. [PMID: 28912891 PMCID: PMC5596439 DOI: 10.7150/thno.19017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/24/2017] [Indexed: 12/12/2022] Open
Abstract
The immune system protects the body against a wide range of infectious diseases and cancer by leveraging the efficiency of immune cells and lymphoid organs. Over the past decade, immune cell/organ therapies based on the manipulation, infusion, and implantation of autologous or allogeneic immune cells/organs into patients have been widely tested and have made great progress in clinical applications. Despite these advances, therapy with natural immune cells or lymphoid organs is relatively expensive and time-consuming. Alternatively, biomimetic materials and strategies have been applied to develop artificial immune cells and lymphoid organs, which have attracted considerable attentions. In this review, we survey the latest studies on engineering biomimetic materials for immunotherapy, focusing on the perspectives of bioengineering artificial antigen presenting cells and lymphoid organs. The opportunities and challenges of this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hunter N. Bomba
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
35
|
Wang C, Ye Y, Hu Q, Bellotti A, Gu Z. Tailoring Biomaterials for Cancer Immunotherapy: Emerging Trends and Future Outlook. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28556553 DOI: 10.1002/adma.201606036] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/04/2017] [Indexed: 05/05/2023]
Abstract
Cancer immunotherapy, as a paradigm shift in cancer treatment, has recently received tremendous attention. The active cancer vaccination, immune checkpoint blockage (ICB) and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer are among these developments that have achieved a significant increase in patient survival in clinical trials. Despite these advancements, emerging research at the interdisciplinary interface of cancer biology, immunology, bioengineering, and materials science is important to further enhance the therapeutic benefits and reduce side effects. Here, an overview of the latest studies on engineering biomaterials for the enhancement of anticancer immunity is given, including the perspectives of delivery of immunomodulatory therapeutics, engineering immune cells, and constructing immune-modulating scaffolds. The opportunities and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
36
|
Abstract
Background Immunotherapy consists of activating the patient’s immune system to fight cancer and has the great potential of preventing future relapses thanks to immunological memory. A great variety of strategies have emerged to harness the immune system against tumors, from the administration of immunomodulatory agents that activate immune cells, to therapeutic vaccines or infusion of previously activated cancer-specific T cells. However, despite great recent progress many difficulties still remain, which prevent the widespread use of immunotherapy. Some of these limitations include: systemic toxicity, weak immune cellular responses or persistence over time and most ultimately costly and time-consuming procedures. Main body Synthetic and natural biomaterials hold great potential to address these hurdles providing biocompatible systems capable of targeted local delivery, co-delivery, and controlled and/or sustained release. In this review we discuss some of the bioengineered solutions and approaches developed so far and how biomaterials can be further implemented to help and shape the future of cancer immunotherapy. Conclusion The bioengineering strategies here presented constitute a powerful toolkit to develop safe and successful novel cancer immunotherapies.
Collapse
|
37
|
Ben-Akiva E, Meyer RA, Wilson DR, Green JJ. Surface engineering for lymphocyte programming. Adv Drug Deliv Rev 2017; 114:102-115. [PMID: 28501510 PMCID: PMC5688954 DOI: 10.1016/j.addr.2017.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
Abstract
The once nascent field of immunoengineering has recently blossomed to include approaches to deliver and present biomolecules to program diverse populations of lymphocytes to fight disease. Building upon improved understanding of the molecular and physical mechanics of lymphocyte activation, varied strategies for engineering surfaces to activate and deactivate T-Cells, B-Cells and natural killer cells are in preclinical and clinical development. Surfaces have been engineered at the molecular level in terms of the presence of specific biological factors, their arrangement on a surface, and their diffusivity to elicit specific lymphocyte fates. In addition, the physical and mechanical characteristics of the surface including shape, anisotropy, and rigidity of particles for lymphocyte activation have been fine-tuned. Utilizing these strategies, acellular systems have been engineered for the expansion of T-Cells and natural killer cells to clinically relevant levels for cancer therapies as well as engineered to program B-Cells to better combat infectious diseases.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Randall A Meyer
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Johns Hopkins Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
38
|
Delcassian D, Sattler S, Dunlop IE. T cell immunoengineering with advanced biomaterials. Integr Biol (Camb) 2017; 9:211-222. [PMID: 28252135 PMCID: PMC6034443 DOI: 10.1039/c6ib00233a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/15/2017] [Indexed: 12/25/2022]
Abstract
Recent advances in biomaterials design offer the potential to actively control immune cell activation and behaviour. Many human diseases, such as infections, cancer, and autoimmune disorders, are partly mediated by inappropriate or insufficient activation of the immune system. T cells play a central role in the host immune response to these diseases, and so constitute a promising cell type for manipulation. In vivo, T cells are stimulated by antigen presenting cells (APC), therefore to design immunoengineering biomaterials that control T cell behaviour, artificial interfaces that mimic the natural APC-T cell interaction are required. This review draws together research in the design and fabrication of such biomaterial interfaces, and highlights efforts to elucidate key parameters in T cell activation, such as substrate mechanical properties and spatial organization of receptors, illustrating how they can be manipulated by bioengineering approaches to alter T cell function.
Collapse
Affiliation(s)
- Derfogail Delcassian
- School of Pharmacy, University of Nottingham, NG7 2RD, UK. and Koch Institute for Integrative Cancer Research, MIT, Massachusetts, 02139, USA
| | - Susanne Sattler
- Imperial College London National Heart and Lung Institute, Du Cane Road, W12 0NN, London, UK
| | - Iain E Dunlop
- Department of Materials, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
39
|
Abstract
This review focuses on summarizing the existing work about nanomaterial-based cancer immunotherapy in detail.
Collapse
Affiliation(s)
- Lijia Luo
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Rui Shu
- University of Chinese Academy of Sciences
- Beijing 100049
- China
- Key Laboratory of Marine Materials and Related Technology
- CAS & Ningbo Institute of Materials Technology and Engineering
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
40
|
ATZIN-MÉNDEZ J, LÓPEZ-GONZÁLEZ J, BÁEZ R, ARENAS-DEL ANGEL M, MONTAÑO L, SILVA-ADAYA D, LASCURAIN R, GOROCICA P. Expansion of quiescent lung adenocarcinoma CD8+ T cells by MUC1-8-mer peptide-T2 cell-β2 microglobulin complexes. Oncol Rep 2016; 35:33-42. [PMID: 26498650 PMCID: PMC4699617 DOI: 10.3892/or.2015.4328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/21/2015] [Indexed: 12/20/2022] Open
Abstract
Adoptive immunotherapy requires the isolation of CD8+ T cells specific for tumor-associated antigens, their expansion in vitro and their transfusion to the patient to mediate a therapeutic effect. MUC1 is an important adenocarcinoma antigen immunogenic for T cells. The MUC1-derived SAPDTRPA (MUC1-8-mer) peptide is a potent epitope recognized by CD8+ T cells in murine models. Likewise, the T2 cell line has been used as an antigen-presenting cell to activate CD8+ T cells, but so far MUC1 has not been assessed in this context. We evaluated whether the MUC1-8-mer peptide can be presented by T2 cells to expand CD25+CD8+ T cells isolated from HLA-A2+ lung adenocarcinoma patients with stage III or IV tumors. The results showed that MUC1-8-mer peptide-loaded T2 cells activated CD8+ T cells from cancer HLA-A2+ patients when anti-CD2, anti-CD28 antibodies and IL-2 were added. The percentage of CD25+CD8+ T cells was 3-fold higher than those in the non-stimulated cells (P=0.018). HLA-A2+ patient cells showed a significant difference (2.3-fold higher) in activation status than HLA-A2+ healthy control cells (P=0.04). Moreover, 77.6% of MUC1-8-mer peptide-specific CD8+ T cells proliferated following a second stimulation with MUC1-8-mer peptide-loaded T2 cells after 10 days of cell culture. There were significant differences in the percentage of basal CD25+CD8+ T cells in relation to the cancer stage; this difference disappeared after MUC1-8-mer peptide stimulation. In conclusion, expansion of CD25+CD8+ T cells by MUC1-8 peptide-loaded T2 cells plus costimulatory signals via CD2, CD28 and IL-2 can be useful in adoptive immunotherapy.
Collapse
Affiliation(s)
- J.A. ATZIN-MÉNDEZ
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - J.S. LÓPEZ-GONZÁLEZ
- Lung Cancer Laboratory, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - R. BÁEZ
- Clinical Oncology and Pneumology, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| | - M.C. ARENAS-DEL ANGEL
- Department of Biochemistry, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - L.F. MONTAÑO
- Immunobiology Laboratory, Department of Cell and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - D. SILVA-ADAYA
- Experimental Laboratory for Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Mexico, DF 14269, Mexico
| | - R. LASCURAIN
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
- Department of Biochemistry, National Autonomous University of Mexico, Mexico, DF 04510, Mexico
| | - P. GOROCICA
- Department of Research in Biochemistry, National Institute of Respiratory Diseases 'Ismael Cosio Villegas', Mexico, DF 14080, Mexico
| |
Collapse
|
41
|
Abstract
The microenvironment is increasingly recognized to have key roles in cancer, and biomaterials provide a means to engineer microenvironments both in vitro and in vivo to study and manipulate cancer. In vitro cancer models using 3D matrices recapitulate key elements of the tumour microenvironment and have revealed new aspects of cancer biology. Cancer vaccines based on some of the same biomaterials have, in parallel, allowed for the engineering of durable prophylactic and therapeutic anticancer activity in preclinical studies, and some of these vaccines have moved to clinical trials. The impact of biomaterials engineering on cancer treatment is expected to further increase in importance in the years to come.
Collapse
Affiliation(s)
- Luo Gu
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - David J Mooney
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
42
|
How to Quantify Penile Corpus Cavernosum Structures with Histomorphometry: Comparison of Two Methods. BIOMED RESEARCH INTERNATIONAL 2015; 2015:832156. [PMID: 26413547 PMCID: PMC4564595 DOI: 10.1155/2015/832156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/31/2015] [Accepted: 04/07/2015] [Indexed: 12/20/2022]
Abstract
The use of morphometrical tools in biomedical research permits the accurate comparison of specimens subjected to different conditions, and the surface density of structures is commonly used for this purpose. The traditional point-counting method is reliable but time-consuming, with computer-aided methods being proposed as an alternative. The aim of this study was to compare the surface density data of penile corpus cavernosum trabecular smooth muscle in different groups of rats, measured by two observers using the point-counting or color-based segmentation method. Ten normotensive and 10 hypertensive male rats were used in this study. Rat penises were processed to obtain smooth muscle immunostained histological slices and photomicrographs captured for analysis. The smooth muscle surface density was measured in both groups by two different observers by the point-counting
method and by the color-based segmentation method. Hypertensive rats showed an increase in smooth muscle surface density by the two methods, and no difference was found between the results of the two observers. However, surface density values were higher by the point-counting method. The use of either method did not influence the final interpretation of the results, and both proved to have adequate reproducibility. However, as differences were found between the two methods, results obtained by either method should not be compared.
Collapse
|
43
|
Meyer RA, Sunshine JC, Green JJ. Biomimetic particles as therapeutics. Trends Biotechnol 2015; 33:514-524. [PMID: 26277289 DOI: 10.1016/j.tibtech.2015.07.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/03/2015] [Accepted: 07/07/2015] [Indexed: 11/28/2022]
Abstract
In recent years, there have been major advances in the development of novel nanoparticle- and microparticle-based therapeutics. An emerging paradigm is the incorporation of biomimetic features into these synthetic therapeutic constructs to enable them to better interface with biological systems. Through the control of size, shape, and material consistency, particle cores have been generated that better mimic natural cells and viruses. In addition, there have been significant advances in biomimetic surface functionalization of particles through the integration of bio-inspired artificial cell membranes and naturally derived cell membranes. Biomimetic technologies enable therapeutic particles to have increased potency to benefit human health.
Collapse
Affiliation(s)
- Randall A Meyer
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith 5017, Baltimore MD, 21231, USA
| | - Joel C Sunshine
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith 5017, Baltimore MD, 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, and Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith 5017, Baltimore MD, 21231, USA.,Departments of Materials Science and Engineering, Oncology, Ophthalmology, and Neurosurgery, Johns Hopkins School of Medicine, 400 N Broadway, Smith 5017, Baltimore MD, 21231, USA
| |
Collapse
|