1
|
Ariail E, Garcia Espinoza N, Stephenson AC, Spangler JB. Emerging approaches for T cell-stimulating platform development. Cell Syst 2024; 15:1198-1208. [PMID: 39701036 DOI: 10.1016/j.cels.2024.11.007] [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: 04/02/2024] [Revised: 06/03/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024]
Abstract
T cells are key mediators of the adaptive immune response, playing both direct and supporting roles in the destruction of foreign pathogenic threats as well as pathologically transformed host cells. The natural process through which T cells are activated requires coordinated molecular interactions between antigen-presenting cells and T cells. Promising advances in biomaterial design have catalyzed the development of artificial platforms that mimic the natural process of T cell stimulation, both to bolster the performance of cell therapies by activating T cells ex vivo prior to adoptive cell transfer and to directly activate T cells in vivo as off-the-shelf treatments. This review focuses on innovative strategies in T cell-stimulating platform design for applications in cancer therapy. We specifically highlight progress in bead-based artificial antigen-presenting cell engineering, hydrogel-based scaffolds, DNA-based systems, alternative polymeric strategies, and soluble activation approaches. Collectively, these advances are expanding the repertoire of tools for targeted immune activation.
Collapse
Affiliation(s)
- Emily Ariail
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nikol Garcia Espinoza
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - A Carson Stephenson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Molecular Microbiology & Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
2
|
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
|
3
|
Shah RK, Cygan E, Kozlik T, Colina A, Zamora AE. Utilizing immunogenomic approaches to prioritize targetable neoantigens for personalized cancer immunotherapy. Front Immunol 2023; 14:1301100. [PMID: 38149253 PMCID: PMC10749952 DOI: 10.3389/fimmu.2023.1301100] [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: 09/24/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Advancements in sequencing technologies and bioinformatics algorithms have expanded our ability to identify tumor-specific somatic mutation-derived antigens (neoantigens). While recent studies have shown neoantigens to be compelling targets for cancer immunotherapy due to their foreign nature and high immunogenicity, the need for increasingly accurate and cost-effective approaches to rapidly identify neoantigens remains a challenging task, but essential for successful cancer immunotherapy. Currently, gene expression analysis and algorithms for variant calling can be used to generate lists of mutational profiles across patients, but more care is needed to curate these lists and prioritize the candidate neoantigens most capable of inducing an immune response. A growing amount of evidence suggests that only a handful of somatic mutations predicted by mutational profiling approaches act as immunogenic neoantigens. Hence, unbiased screening of all candidate neoantigens predicted by Whole Genome Sequencing/Whole Exome Sequencing may be necessary to more comprehensively access the full spectrum of immunogenic neoepitopes. Once putative cancer neoantigens are identified, one of the largest bottlenecks in translating these neoantigens into actionable targets for cell-based therapies is identifying the cognate T cell receptors (TCRs) capable of recognizing these neoantigens. While many TCR-directed screening and validation assays have utilized bulk samples in the past, there has been a recent surge in the number of single-cell assays that provide a more granular understanding of the factors governing TCR-pMHC interactions. The goal of this review is to provide an overview of existing strategies to identify candidate neoantigens using genomics-based approaches and methods for assessing neoantigen immunogenicity. Additionally, applications, prospects, and limitations of some of the current single-cell technologies will be discussed. Finally, we will briefly summarize some of the recent models that have been used to predict TCR antigen specificity and analyze the TCR receptor repertoire.
Collapse
Affiliation(s)
- Ravi K. Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Cygan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tanya Kozlik
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Alfredo Colina
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anthony E. Zamora
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
4
|
Mateus D, Sebastião AI, Frasco MF, Carrascal MA, Falcão A, Gomes CM, Neves B, Sales MGF, Cruz MT. Artificial Dendritic Cells: A New Era of Promising Antitumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303940. [PMID: 37469192 DOI: 10.1002/smll.202303940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/03/2023] [Indexed: 07/21/2023]
Abstract
The accelerated development of antitumor immunotherapies in recent years has brought immunomodulation into the spotlight. These include immunotherapeutic treatments with dendritic cell (DC)-based vaccines which can elicit tumor-specific immune responses and prolong survival. However, this personalized treatment has several drawbacks, including being costly, labor-intensive, and time consuming. This has sparked interest in producing artificial dendritic cells (aDCs) to open up the possibility of standardized "off-the-shelf" protocols and circumvent the cumbersome and expensive personalized medicine. aDCs take advantage of materials that can be designed and tailored for specific clinical applications. Here, an overview of the immunobiology underlying antigen presentation by DCs is provided in an attempt to select the key features to be mimicked and/or improved through the development of aDCs. The inherent properties of aDCs that greatly impact their performance in vivo and, consequently, the fate of the triggered immune response are also outlined.
Collapse
Affiliation(s)
- Daniela Mateus
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | - Ana I Sebastião
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Manuela F Frasco
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | | | - Amílcar Falcão
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Célia M Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Bruno Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Maria G F Sales
- BioMark@UC/CEB - LABBELS Department of Chemical Engineering, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3030-790, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy of the University of Coimbra, Coimbra, 3000-548, Portugal
- Center for Neuroscience and Cell Biology-CNC, University of Coimbra, Coimbra, 3004-504, Portugal
| |
Collapse
|
5
|
Ben-Akiva E, Hickey JW, Meyer RA, Isser A, Shannon SR, Livingston NK, Rhodes KR, Kosmides AK, Warren TR, Tzeng SY, Schneck JP, Green JJ. Shape matters: Biodegradable anisotropic nanoparticle artificial antigen presenting cells for cancer immunotherapy. Acta Biomater 2023; 160:187-197. [PMID: 36812956 PMCID: PMC10335041 DOI: 10.1016/j.actbio.2023.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
Artificial antigen presenting cells are biomimetic particles that recapitulate the signals presented by natural antigen presenting cells in order to stimulate T cells in an antigen-specific manner using an acellular platform. We have engineered an enhanced nanoscale biodegradable artificial antigen presenting cell by modulating particle shape to achieve a nanoparticle geometry that allows for increased radius of curvature and surface area for T cell contact. The non-spherical nanoparticle artificial antigen presenting cells developed here have reduced nonspecific uptake and improved circulation time compared both to spherical nanoparticles and to traditional microparticle technologies. Additionally, the anisotropic nanoparticle artificial antigen presenting cells efficiently engage with and activate T cells, ultimately leading to a marked anti-tumor effect in a mouse melanoma model that their spherical counterparts were unable to achieve. STATEMENT OF SIGNIFICANCE: Artificial antigen presenting cells (aAPC) can activate antigen-specific CD8+ T cells but have largely been limited to microparticle-based platforms and ex vivo T cell expansion. Although more amenable to in vivo use, nanoscale aAPC have traditionally been ineffective due to limited surface area available for T cell interaction. In this work, we engineered non-spherical biodegradable nanoscale aAPC to investigate the role of particle geometry and develop a translatable platform for T cell activation. The non-spherical aAPC developed here have increased surface area and a flatter surface for T cell engagement and, therefore, can more effectively stimulate antigen-specific T cells, resulting in anti-tumor efficacy in a mouse melanoma model.
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
| | - John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, 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
| | - Ariel Isser
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sydney R Shannon
- 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; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Natalie K Livingston
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kelly R Rhodes
- 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
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tiarra R Warren
- 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
| | - Stephany Y Tzeng
- 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
| | - Jonathan P Schneck
- Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Medicine, 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; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
6
|
Lee-Chang C, Lesniak MS. Next-generation antigen-presenting cell immune therapeutics for gliomas. J Clin Invest 2023; 133:e163449. [PMID: 36719372 PMCID: PMC9888388 DOI: 10.1172/jci163449] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Antigen presentation machinery and professional antigen-presenting cells (APCs) are fundamental for an efficacious immune response against cancers, especially in the context of T cell-centric immunotherapy. Dendritic cells (DCs), the gold standard APCs, play a crucial role in initiating and maintaining a productive antigen-specific adaptive immunity. In recent decades, ex vivo-differentiated DCs from circulating CD14+ monocytes have become the reference for APC-based immunotherapy. DCs loaded with tumor-associated antigens, synthetic peptides, or RNA activate T cells with antitumor properties. This strategy has paved the way for the development of alternative antigen-presenting vaccination strategies, such as monocytes, B cells, and artificial APCs, that have shown effective therapeutic outcomes in preclinical cancer models. The search for alternative APC platforms was initiated by the overall limited clinical impact of DC vaccines, especially in indications such as gliomas, a primary brain tumor known for resistance to any immune intervention. In this Review, we navigate the APC immune therapeutics' past, present, and future in the context of primary brain tumors.
Collapse
Affiliation(s)
- Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Malnati Brain Tumor Institute, Chicago, Illinois, USA
| |
Collapse
|
7
|
Harari-Steinfeld R, Abhinav Ayyadevara VSS, Cuevas L, Marincola F, Roh KH. Standardized in-vitro evaluation of CAR-T cells using acellular artificial target particles. Front Immunol 2022; 13:994532. [PMID: 36341361 PMCID: PMC9632174 DOI: 10.3389/fimmu.2022.994532] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/26/2022] [Indexed: 10/31/2024] Open
Abstract
The horizon of immunotherapy using CAR-T cells is continuously extending to treat solid tumors beyond the success in the treatment of liquid tumors. Precise in-vitro evaluations of CAR-T cells for their phenotypes, quantity and quality of activation in various tumor microenvironments including different antigen densities, and the resulting effector functions are critical for the successful development of CAR-T therapies and safe translation to clinics. Unfortunately, the development of methods and tools to accommodate these needs have been lagging behind. Here, we developed a novel biomaterial platform, acellular artificial target particles (aaTPs) against CAR-T cells, using magnetic microbeads that are already widely employed in the manufacturing of T cell products. By devising a simple and standardized procedure, we precisely controlled the antigen surface densities presented on the aaTPs for a wide range. By co-incubation of aaTPs with CAR-T cells followed by flow cytometry and cytokine assays, we quantitatively determined the antigen-specific and dose-dependent activation of anti-HER2 CAR-T cells. We also demonstrated that the aaTP can serve as a clean target cell in in-vitro assays to prove the proposed mechanism of action of a next-generation CAR-T product. Overall, the simple, inexpensive, modular and precisely controllable synthetic nature of aaTPs enables the development of clean and standardized in-vitro assays for CAR-T cells, which provides critical advantages over the conventional assays using target cell lines. The design of aaTPs can be extended to include other tumor antigens and relevant surface molecules of physiological target cells. Thus, the aaTP platform has great potential as a standardized tool for the development and evaluation of both conventional and new CAR-T products in the context of approval from regulatory agencies and clinical translation.
Collapse
Affiliation(s)
| | - V. S. S. Abhinav Ayyadevara
- Biotechnology Science and Engineering, The University of Alabama in Huntsville, Huntsville, AL, United States
| | | | | | - Kyung-Ho Roh
- Biotechnology Science and Engineering, The University of Alabama in Huntsville, Huntsville, AL, United States
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, United States
| |
Collapse
|
8
|
Sung BY, Lin YH, Kong Q, Shah PD, Glick Bieler J, Palmer S, Weinhold KJ, Chang HR, Huang H, Avery RK, Schneck J, Chiu YL. Wnt activation promotes memory T cell polyfunctionality via epigenetic regulator PRMT1. J Clin Invest 2022; 132:e140508. [PMID: 35040433 PMCID: PMC8759796 DOI: 10.1172/jci140508] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
T cell polyfunctionality is a hallmark of protective immunity against pathogens and cancer, yet the molecular mechanism governing it remains mostly elusive. We found that canonical Wnt agonists inhibited human memory CD8+ T cell differentiation while simultaneously promoting the generation of highly polyfunctional cells. Downstream effects of Wnt activation persisted after removal of the drug, and T cells remained polyfunctional following subsequent cell division, indicating the effect is epigenetically regulated. Wnt activation induced a gene expression pattern that is enriched with stem cell-specific gene signatures and upregulation of protein arginine methyltransferase 1 (PRMT1), a known epigenetic regulator. PRMT1+CD8+ T cells are associated with enhanced polyfunctionality, especially the ability to produce IL-2. In contrast, inhibition of PRMT1 ameliorated the effects of Wnt on polyfunctionality. Chromatin immunoprecipitation revealed that H4R3me2a, a permissive transcription marker mediated by PRMT1, increased at the IL-2 promoter loci following Wnt activation. In vivo, Wnt-treated T cells exhibited superior polyfunctionality and persistence. When applied to cytomegalovirus (CMV) donor-seropositive, recipient-seronegative patients (D+/R-) lung transplant patient samples, Wnt activation enhanced CMV-specific T cell polyfunctionality, which is important in controlling CMV diseases. These findings reveal a molecular mechanism governing T cell polyfunctionality and identify PRMT1 as a potential target for T cell immunotherapy.
Collapse
Affiliation(s)
- Bo-Yi Sung
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Microbiology and Immunology
- Department of Biomedical Engineering, and
| | - Yi-Hsin Lin
- Department of Biomedical Engineering, and
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | - Pali D. Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joan Glick Bieler
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Kent J. Weinhold
- Department of Surgery, and Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robin K. Avery
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan Schneck
- Institute of Cell Engineering and
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine and Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland. USA
| | - Yen-Ling Chiu
- Institute of Cell Engineering and
- Graduate Institute of Medicine and Graduate Program in Biomedical Informatics, Yuan Ze University, Taoyuan, Taiwan
- Department of Medical Research, Far Eastern Memorial Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
9
|
Lin JC, Hsu CY, Chen JY, Fang ZS, Chen HW, Yao BY, Shiau GHM, Tsai JS, Gu M, Jung M, Lee TY, Hu CMJ. Facile Transformation of Murine and Human Primary Dendritic Cells into Robust and Modular Artificial Antigen-Presenting Systems by Intracellular Hydrogelation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101190. [PMID: 34096117 DOI: 10.1002/adma.202101190] [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] [Received: 02/10/2021] [Revised: 04/01/2021] [Indexed: 06/12/2023]
Abstract
The growing enthusiasm for cancer immunotherapies and adoptive cell therapies has prompted increasing interest in biomaterials development mimicking natural antigen-presenting cells (APCs) for T-cell expansion. In contrast to conventional bottom-up approaches aimed at layering synthetic substrates with T-cell activation cues, transformation of live dendritic cells (DCs) into artificial APCs (aAPCs) is demonstrated herein using a facile and minimally disruptive hydrogelation technique. Through direct intracellular permeation of poly(ethylene glycol) diacrylate (PEG-DA) hydrogel monomer and UV-activated radical polymerization, intracellular hydrogelation is rapidly accomplished on DCs with minimal influence on cellular morphology and surface antigen display, yielding highly robust and modular cell-gel hybrid constructs amenable to peptide antigen exchange, storable by freezing and lyophilization, and functionalizable with cytokine-releasing carriers for T-cell modulation. The DC-derived aAPCs are shown to induce prolonged T-cell expansion and improve anticancer efficacy of adoptive T-cell therapy in mice compared to nonexpanded control T cells, and the gelation technique is further demonstrated to stabilize primary DCs derived from human donors. The work presents a versatile approach for generating a new class of cell-mimicking biomaterials and opens new venues for immunological interrogation and immunoengineering.
Collapse
Affiliation(s)
- Jung-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Chung-Yao Hsu
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Jui-Yi Chen
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Zih-Syun Fang
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Road, Da'an District, Taipei, Taiwan, 106, Republic of China
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Gwo Harn M Shiau
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Jeng-Shiang Tsai
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| | - Ming Gu
- Celtec Inc., One Broadway, Cambridge, MA, 02142, USA
- Celtec Inc., 15-7F, No 99, Sec 1, Xintai 5th Road, New Taipei City, Taiwan, 22175, Republic of China
| | - Meiying Jung
- Celtec Inc., One Broadway, Cambridge, MA, 02142, USA
- Celtec Inc., 15-7F, No 99, Sec 1, Xintai 5th Road, New Taipei City, Taiwan, 22175, Republic of China
| | - Tong-Young Lee
- Celtec Inc., One Broadway, Cambridge, MA, 02142, USA
- Celtec Inc., 15-7F, No 99, Sec 1, Xintai 5th Road, New Taipei City, Taiwan, 22175, Republic of China
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, Taiwan, 115, Republic of China
| |
Collapse
|
10
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
11
|
Carreira B, Acúrcio RC, Matos AI, Peres C, Pozzi S, Vaskovich‐Koubi D, Kleiner R, Bento M, Satchi‐Fainaro R, Florindo HF. Nanomedicines as Multifunctional Modulators of Melanoma Immune Microenvironment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ana I. Matos
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Mariana Bento
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| |
Collapse
|
12
|
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]
|
13
|
Ruan H, Bu L, Hu Q, Cheng H, Lu W, Gu Z. Strategies of Combination Drug Delivery for Immune Checkpoint Blockades. Adv Healthc Mater 2019; 8:e1801099. [PMID: 30548835 DOI: 10.1002/adhm.201801099] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/29/2018] [Indexed: 12/19/2022]
Abstract
The past few years have witnessed vast clinical accomplishments of immune checkpoint blockades (ICB), which block the regulatory receptor expressed on immune cells or tumor cells to prevent the suppression of antitumor cytotoxic T-cell responses. Despite this, limitations still exist, such as low objective response rate (ORR) and the risk of immune-related side effects. To address these issues, combination treatment strategies are vastly explored and recommended. This review summarizes recent advances in combination of ICB with therapies that participate in different stages of cancer immune cycle, including tumor antigen release, tumor antigen presentation, T-cell activation, recognition of cancer cells by T-cells, and tumor-killing activity. Challenges and potential opportunities of combination approaches in this field are also discussed.
Collapse
Affiliation(s)
- Huitong Ruan
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryFudan University Shanghai 201203 China
- Department of BioengineeringUniversity of California Los Angeles CA 90095 USA
- California NanoSystems InstituteJonsson Comprehensive Cancer Center and Center for Minimally Invasive TherapeuticsUniversity of California Los Angeles CA 90095 USA
- Department of Materials Science & EngineeringDrexel University Philadelphia PA 19104 USA
| | - Linlin Bu
- Department of BioengineeringUniversity of California Los Angeles CA 90095 USA
- California NanoSystems InstituteJonsson Comprehensive Cancer Center and Center for Minimally Invasive TherapeuticsUniversity of California Los Angeles CA 90095 USA
| | - Quanyin Hu
- Department of BioengineeringUniversity of California Los Angeles CA 90095 USA
- California NanoSystems InstituteJonsson Comprehensive Cancer Center and Center for Minimally Invasive TherapeuticsUniversity of California Los Angeles CA 90095 USA
| | - Hao Cheng
- Department of Materials Science & EngineeringDrexel University Philadelphia PA 19104 USA
| | - Weiyue Lu
- Department of PharmaceuticsSchool of PharmacyKey Laboratory of Smart Drug DeliveryFudan University Shanghai 201203 China
| | - Zhen Gu
- Department of BioengineeringUniversity of California Los Angeles CA 90095 USA
- California NanoSystems InstituteJonsson Comprehensive Cancer Center and Center for Minimally Invasive TherapeuticsUniversity of California Los Angeles CA 90095 USA
| |
Collapse
|
14
|
Hickey JW, Isser AY, Vicente FP, Warner SB, Mao HQ, Schneck JP. Efficient magnetic enrichment of antigen-specific T cells by engineering particle properties. Biomaterials 2018; 187:105-116. [PMID: 30312851 DOI: 10.1016/j.biomaterials.2018.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/08/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022]
Abstract
Magnetic particles can enrich desired cell populations to aid in understanding cell-type functions and mechanisms, diagnosis, and therapy. As cells are heterogeneous in ligand type, location, expression, and density, careful consideration of magnetic particle design for positive isolation is necessary. Antigen-specific immune cells have low frequencies, which has made studying, identifying, and utilizing these cells for therapy a challenge. Here we demonstrate the importance of magnetic particle design based on the biology of T cells. We create magnetic particles which recognize rare antigen-specific T cells and quantitatively investigate important particle properties including size, concentration, ligand density, and ligand choice in enriching these rare cells. We observe competing optima among particle parameters, with 300 nm particles functionalized with a high density of antigen-specific ligand achieving the highest enrichment and recovery of target cells. In enriching and then activating an endogenous response, 300 nm aAPCs generate nearly 65% antigen-specific T cells with at least 450-fold expansion from endogenous precursors and a 5-fold increase in numbers of antigen-specific cells after only seven days. This systematic study of particle properties in magnetic enrichment provides a case study for the engineering design principles of particles for the isolation of rare cells through biological ligands.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, School of Medicine, Baltimore, MD, USA; Translational Tissue Engineering Center, Baltimore, MD, USA; Institute for Nanobiotechnology, Baltimore, MD, USA
| | - Ariel Y Isser
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, School of Medicine, Baltimore, MD, USA
| | - Fernando P Vicente
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, USA
| | - Samuel B Warner
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, Baltimore, MD, USA; Institute for Nanobiotechnology, Baltimore, MD, USA; Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, USA
| | - Jonathan P Schneck
- Institute for Cell Engineering, School of Medicine, Baltimore, MD, USA; Department of Pathology, School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Baltimore, MD, USA; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Truong DH, Tran TTP, Nguyen HT, Phung CD, Pham TT, Yong CS, Kim JO, Tran TH. Modulating T-cell-based cancer immunotherapy via particulate systems. J Drug Target 2018; 27:145-163. [PMID: 29741964 DOI: 10.1080/1061186x.2018.1474360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunotherapy holds tremendous promise for improving cancer treatment in which an appropriate stimulator may naturally trigger the immune system to control cancer. Up-to-date, adoptive T-cell therapy has received two new FDA approvals that provide great hope for some cancer patient groups. Nevertheless, expense and safety-related issues require further study to obtain insight into targets for efficient immunotherapy. The development of material science was largely responsible for providing a promising horizon to strengthen immunoengineering. In this review, we focus on T-cell characteristics in the context of the immune system against cancer and discuss several approaches of exploiting engineered particles to manipulate the responses of T cells and the tumour microenvironment.
Collapse
Affiliation(s)
- Duy Hieu Truong
- a Institute of Research and Development, Duy Tan University , Da Nang , Vietnam
| | - Thi Thu Phuong Tran
- b The Institute of Molecular Genetics of Montpellier, CNRS , Montpellier , France
| | - Hanh Thuy Nguyen
- c College of Pharmacy , Yeungnam University , Gyeongsan , Republic of Korea
| | - Cao Dai Phung
- c College of Pharmacy , Yeungnam University , Gyeongsan , Republic of Korea
| | - Tung Thanh Pham
- c College of Pharmacy , Yeungnam University , Gyeongsan , Republic of Korea
| | - Chul Soon Yong
- c College of Pharmacy , Yeungnam University , Gyeongsan , Republic of Korea
| | - Jong Oh Kim
- c College of Pharmacy , Yeungnam University , Gyeongsan , Republic of Korea
| | - Tuan Hiep Tran
- d Department for Management of Science and Technology Development , Ton Duc Thang University , Ho Chi Minh City , Vietnam.,e Faculty of Pharmacy , Ton Duc Thang University , Ho Chi Minh City , Vietnam
| |
Collapse
|
16
|
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
|
17
|
Schütz C, Varela JC, Perica K, Haupt C, Oelke M, Schneck JP. Antigen-specific T cell Redirectors: a nanoparticle based approach for redirecting T cells. Oncotarget 2018; 7:68503-68512. [PMID: 27602488 PMCID: PMC5356569 DOI: 10.18632/oncotarget.11785] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/27/2016] [Indexed: 12/20/2022] Open
Abstract
Redirection of T cells to target and destroy tumors has become an important clinical tool and major area of research in tumor immunology. Here we present a novel, nanoparticle-based approach to selectively bind antigen-specific cytotoxic T cells (CTL) and redirect them to kill tumors, termed ATR (Antigen-specific T cell Redirectors). ATR were generated by decorating nanoparticles with both an antigen-specific T cell binding moiety, either peptide loaded MHC-Ig dimer or clonotypic anti-TCR antibody, and a model tumor cell binding moiety, anti-CD19 antibody to engage CD19+ tumor cells. ATR stably bind tumor cells and CTL in a dose dependent fashion and stimulate antigen-specific conjugate formation between those cells. ATR induced redirected lysis of tumor cells in vitro, as demonstrated by 51Cr-release killing. In vivo ATR administration led to reduced tumor growth in a SCID/beige human lymphoma treatment model. In summary, ATR represent a novel, nanoparticle based approach for redirecting antigen-specific CTL to kill tumors.
Collapse
Affiliation(s)
- Christian Schütz
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Current address: Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Juan Carlos Varela
- Division of Hematology, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Karlo Perica
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Carl Haupt
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Mathias Oelke
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,NexImmune Inc., Gaithersburg, Maryland, USA
| | - Jonathan P Schneck
- Institute of Cell Engineering and Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Artificial Methods for T Cell Activation: Critical Tools in T Cell Biology and T Cell Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:207-219. [PMID: 30471035 DOI: 10.1007/978-981-13-0445-3_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Antigen-specific immunity conferred by T lymphocytes is a result of complex molecular interactions at the immunological synapse. A variety of biomimetic approaches have been devised to artificially induce T cell activation either to study the T cell biology or to expand and prime the therapeutic T cell populations. Here we first briefly review the molecular and cellular, structural and phenotypical bases that are involved in T cell activation. The artificial methods for T cell activation are then discussed in two grand categories, the soluble (3D) and the surface-anchored (2D) platforms with their design parameters. With the growing number of successful adoptive T cell therapies, the spurring demands for effective and safe T cell expansion as well as precise control over resulting T cell functions and phenotypes warrant the extensions of engineering parameters in the development of novel methodologies for T cell activation.
Collapse
|
19
|
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
|
20
|
Neal LR, Bailey SR, Wyatt MM, Bowers JS, Majchrzak K, Nelson MH, Haupt C, Paulos CM, Varela JC. The Basics of Artificial Antigen Presenting Cells in T Cell-Based Cancer Immunotherapies. JOURNAL OF IMMUNOLOGY RESEARCH AND THERAPY 2017; 2:68-79. [PMID: 28825053 PMCID: PMC5560309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Adoptive T cell transfer (ACT) can mediate objective responses in patients with advanced malignancies. There have been major advances in this field, including the optimization of the ex vivo generation of tumor-reactive lymphocytes to ample numbers for effective ACT therapy via the use of natural and artificial antigen presenting cells (APCs). Herein we review the basic properties of APCs and how they have been manufactured through the years to augment vaccine and T cell-based cancer therapies. We then discuss how these novel APCs impact the function and memory properties of T cells. Finally, we propose new ways to synthesize aAPCs to augment the therapeutic effectiveness of antitumor T cells for ACT therapy.
Collapse
Affiliation(s)
- Lillian R. Neal
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Stefanie R. Bailey
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Megan M. Wyatt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Jacob S. Bowers
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Kinga Majchrzak
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Michelle H. Nelson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Carl Haupt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| | - Chrystal M. Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Dermatological Surgery and Dermatology, Medical University of South Carolina, Charleston, SC 29425
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | - Juan C. Varela
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, 29425
| |
Collapse
|
21
|
Biomimetic biodegradable artificial antigen presenting cells synergize with PD-1 blockade to treat melanoma. Biomaterials 2016; 118:16-26. [PMID: 27940380 DOI: 10.1016/j.biomaterials.2016.11.038] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 01/08/2023]
Abstract
Biomimetic materials that target the immune system and generate an anti-tumor responses hold promise in augmenting cancer immunotherapy. These synthetic materials can be engineered and optimized for their biodegradability, physical parameters such as shape and size, and controlled release of immune-modulators. As these new platforms enter the playing field, it is imperative to understand their interaction with existing immunotherapies since single-targeted approaches have limited efficacy. Here, we investigate the synergy between a PLGA-based artificial antigen presenting cell (aAPC) and a checkpoint blockade molecule, anti-PD1 monoclonal antibody (mAb). The combination of antigen-specific aAPC-based activation and anti-PD-1 mAb checkpoint blockade induced the greatest IFN-γ secretion by CD8+ T cells in vitro. Combination treatment also acted synergistically in an in vivo murine melanoma model to result in delayed tumor growth and extended survival, while either treatment alone had no effect. This was shown mechanistically to be due to decreased PD-1 expression and increased antigen-specific proliferation of CD8+ T cells within the tumor microenvironment and spleen. Thus, biomaterial-based therapy can synergize with other immunotherapies and motivates the translation of biomimetic combinatorial treatments.
Collapse
|
22
|
Perica K, Bieler JG, Schütz C, Varela JC, Douglass J, Skora A, Chiu YL, Oelke M, Kinzler K, Zhou S, Vogelstein B, Schneck JP. Enrichment and Expansion with Nanoscale Artificial Antigen Presenting Cells for Adoptive Immunotherapy. ACS NANO 2015; 9:6861-71. [PMID: 26171764 PMCID: PMC5082131 DOI: 10.1021/acsnano.5b02829] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Adoptive immunotherapy (AIT) can mediate durable regression of cancer, but widespread adoption of AIT is limited by the cost and complexity of generating tumor-specific T cells. Here we develop an Enrichment + Expansion strategy using paramagnetic, nanoscale artificial antigen presenting cells (aAPC) to rapidly expand tumor-specific T cells from rare naïve precursors and predicted neo-epitope responses. Nano-aAPC are capable of enriching rare tumor-specific T cells in a magnetic column and subsequently activating them to induce proliferation. Enrichment + Expansion resulted in greater than 1000-fold expansion of both mouse and human tumor-specific T cells in 1 week, with nano-aAPC based enrichment conferring a proliferation advantage during both in vitro culture and after adoptive transfer in vivo. Robust T cell responses were seen not only for shared tumor antigens, but also for computationally predicted neo-epitopes. Streamlining the rapid generation of large numbers of tumor-specific T cells in a cost-effective fashion through Enrichment + Expansion can be a powerful tool for immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Jacqueline Douglass
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Andrew Skora
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | | | | - Kenneth Kinzler
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Shibin Zhou
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Bert Vogelstein
- ∥Ludwig Cancer Research Center and Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | |
Collapse
|
23
|
Perica K, Kosmides AK, Schneck JP. Linking form to function: Biophysical aspects of artificial antigen presenting cell design. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:781-90. [PMID: 25200637 PMCID: PMC4344884 DOI: 10.1016/j.bbamcr.2014.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/22/2022]
Abstract
Artificial antigen presenting cells (aAPCs) are engineered platforms for T cell activation and expansion, synthesized by coupling T cell activating proteins to the surface of cell lines or biocompatible particles. They can serve both as model systems to study the basic aspects of T cell signaling and translationally as novel approaches for either active or adoptive immunotherapy. Historically, these reductionist systems have not been designed to mimic the temporally and spatially complex interactions observed during endogenous T cell-APC contact, which include receptor organization at both micro- and nanoscales and dynamic changes in cell and membrane morphologies. Here, we review how particle size and shape, as well as heterogenous distribution of T cell activating proteins on the particle surface, are critical aspects of aAPC design. In doing so, we demonstrate how insights derived from endogenous T cell activation can be applied to optimize aAPC, and in turn how aAPC platforms can be used to better understand endogenous T cell stimulation. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan P Schneck
- Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Bruns H, Bessell C, Varela JC, Haupt C, Fang J, Pasemann S, Mackensen A, Oelke M, Schneck JP, Schütz C. CD47 Enhances In Vivo Functionality of Artificial Antigen-Presenting Cells. Clin Cancer Res 2015; 21:2075-83. [PMID: 25593301 DOI: 10.1158/1078-0432.ccr-14-2696] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/04/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE Artificial antigen-presenting cells, aAPC, have successfully been used to stimulate antigen-specific T-cell responses in vitro as well as in vivo. Although aAPC compare favorably with autologous dendritic cells in vitro, their effect in vivo might be diminished through rapid clearance by macrophages. Therefore, to prevent uptake and minimize clearance of aAPC by macrophages, thereby increasing in vivo functionality, we investigated the efficiency of "don't eat me" three-signal aAPC compared with classical two-signal aAPC. EXPERIMENTAL DESIGN To generate "don't eat me" aAPC, CD47 was additionally immobilized onto classical aAPC (aAPC(CD47+)). aAPC and aAPC(CD47+) were analyzed in in vitro human primary T-cell and macrophage cocultures. In vivo efficiency was compared in a NOD/SCID T-cell proliferation and a B16-SIY melanoma model. RESULTS This study demonstrates that aAPC(CD47+) in coculture with human macrophages show a CD47 concentration-dependent inhibition of phagocytosis, whereas their ability to generate and expand antigen-specific T cells was not affected. Furthermore, aAPC(CD47+)-generated T cells displayed equivalent killing abilities and polyfunctionality when compared with aAPC-generated T cells. In addition, in vivo studies demonstrated an enhanced stimulatory capacity and tumor inhibition of aAPC(CD47+) over normal aAPC in conjunction with diverging biodistribution in different organs. CONCLUSIONS Our data for the first time show that aAPC functionalized with CD47 maintain their stimulatory capacity in vitro and demonstrate enhanced in vivo efficiency. Thus, these next-generation aAPC(CD47+) have a unique potential to enhance the application of the aAPC technology for future immunotherapy approaches.
Collapse
Affiliation(s)
- Heiko Bruns
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Catherine Bessell
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Juan Carlos Varela
- Division of Hematology, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Carl Haupt
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jerry Fang
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Shirin Pasemann
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Mathias Oelke
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jonathan P Schneck
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Christian Schütz
- Institute of Cell Engineering, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
25
|
Eggermont LJ, Paulis LE, Tel J, Figdor CG. Towards efficient cancer immunotherapy: advances in developing artificial antigen-presenting cells. Trends Biotechnol 2014; 32:456-65. [PMID: 24998519 PMCID: PMC4154451 DOI: 10.1016/j.tibtech.2014.06.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 01/07/2023]
Abstract
Active anti-cancer immune responses depend on efficient presentation of tumor antigens and co-stimulatory signals by antigen-presenting cells (APCs). Therapy with autologous natural APCs is costly and time-consuming and results in variable outcomes in clinical trials. Therefore, development of artificial APCs (aAPCs) has attracted significant interest as an alternative. We discuss the characteristics of various types of acellular aAPCs, and their clinical potential in cancer immunotherapy. The size, shape, and ligand mobility of aAPCs and their presentation of different immunological signals can all have significant effects on cytotoxic T cell activation. Novel optimized aAPCs, combining carefully tuned properties, may lead to efficient immunomodulation and improved clinical responses in cancer immunotherapy.
Collapse
Affiliation(s)
- Loek J Eggermont
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Leonie E Paulis
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jurjen Tel
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud University Medical Centre and Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
van der Weijden J, Paulis LE, Verdoes M, van Hest JCM, Figdor CG. The right touch: design of artificial antigen-presenting cells to stimulate the immune system. Chem Sci 2014. [DOI: 10.1039/c4sc01112k] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
27
|
Perica K, Tu A, Richter A, Bieler JG, Edidin M, Schneck JP. Magnetic field-induced T cell receptor clustering by nanoparticles enhances T cell activation and stimulates antitumor activity. ACS NANO 2014; 8:2252-60. [PMID: 24564881 PMCID: PMC4004316 DOI: 10.1021/nn405520d] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/05/2014] [Indexed: 05/20/2023]
Abstract
Iron-dextran nanoparticles functionalized with T cell activating proteins have been used to study T cell receptor (TCR) signaling. However, nanoparticle triggering of membrane receptors is poorly understood and may be sensitive to physiologically regulated changes in TCR clustering that occur after T cell activation. Nano-aAPC bound 2-fold more TCR on activated T cells, which have clustered TCR, than on naive T cells, resulting in a lower threshold for activation. To enhance T cell activation, a magnetic field was used to drive aggregation of paramagnetic nano-aAPC, resulting in a doubling of TCR cluster size and increased T cell expansion in vitro and after adoptive transfer in vivo. T cells activated by nano-aAPC in a magnetic field inhibited growth of B16 melanoma, showing that this novel approach, using magnetic field-enhanced nano-aAPC stimulation, can generate large numbers of activated antigen-specific T cells and has clinically relevant applications for adoptive immunotherapy.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Institute of Cell Engineering, Department of Biology, Department of Pathology, and Departments of Oncology and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Ang Tu
- Department of Biomedical Engineering, Institute of Cell Engineering, Department of Biology, Department of Pathology, and Departments of Oncology and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | | | - Joan Glick Bieler
- Department of Biomedical Engineering, Institute of Cell Engineering, Department of Biology, Department of Pathology, and Departments of Oncology and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Michael Edidin
- Department of Biomedical Engineering, Institute of Cell Engineering, Department of Biology, Department of Pathology, and Departments of Oncology and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Jonathan P. Schneck
- Department of Biomedical Engineering, Institute of Cell Engineering, Department of Biology, Department of Pathology, and Departments of Oncology and Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
28
|
Sunshine JC, Perica K, Schneck JP, Green JJ. Particle shape dependence of CD8+ T cell activation by artificial antigen presenting cells. Biomaterials 2014; 35:269-277. [PMID: 24099710 PMCID: PMC3902087 DOI: 10.1016/j.biomaterials.2013.09.050] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/14/2013] [Indexed: 12/15/2022]
Abstract
Previous work developing particle-based acellular, artificial antigen presenting cells (aAPCs) has focused exclusively on spherical platforms. To explore the role of shape, we generated ellipsoidal PLGA microparticles with varying aspect ratios (ARs) and synthesized aAPCs from them. The ellipsoidal biomimetic aAPCs with high-AR showed significantly enhanced in vitro and in vivo activity above spherical aAPCs with particle volume and antigen content held constant. Confocal imaging indicates that CD8+ T cells preferentially migrate to and are activated by interaction with the long axis of the aAPC. Importantly, enhanced activity of high-AR aAPCs was seen in a mouse melanoma model, with high-AR aAPCs improving melanoma survival compared to non-cognate aAPCs (p = 0.004) and cognate spherical aAPCs (p = 0.05). These findings indicate that particle geometry is a critical design criterion in the generation of aAPCs, and may offer insight into the essential role of geometry in the interaction between CD8+ T cells and biological APCs.
Collapse
Affiliation(s)
- Joel C. Sunshine
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Jonathan P. Schneck
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| | - Jordan J. Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD. 21231
- Department of Ophthalmology Johns Hopkins University School of Medicine, Baltimore, MD. 21231
| |
Collapse
|
29
|
Perica K, De León Medero A, Durai M, Chiu YL, Bieler JG, Sibener L, Niemöller M, Assenmacher M, Richter A, Edidin M, Oelke M, Schneck J. Nanoscale artificial antigen presenting cells for T cell immunotherapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2014; 10:119-29. [PMID: 23891987 PMCID: PMC4114774 DOI: 10.1016/j.nano.2013.06.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 06/25/2013] [Accepted: 06/30/2013] [Indexed: 10/26/2022]
Abstract
Artificial antigen presenting cells (aAPC), which deliver stimulatory signals to cytotoxic lymphocytes, are a powerful tool for both adoptive and active immunotherapy. Thus far, aAPC have been synthesized by coupling T cell activating proteins such as CD3 or MHC-peptide to micron-sized beads. Nanoscale platforms have different trafficking and biophysical interaction properties and may allow development of new immunotherapeutic strategies. We therefore manufactured aAPC based on two types of nanoscale particle platforms: biocompatible iron-dextran paramagnetic particles (50-100 nm in diameter) and avidin-coated quantum dot nanocrystals (~30 nm). Nanoscale aAPC induced antigen-specific T cell proliferation from mouse splenocytes and human peripheral blood T cells. When injected in vivo, both iron-dextran particles and quantum dot nanocrystals enhanced tumor rejection in a subcutaneous mouse melanoma model. This is the first description of nanoscale aAPC that induce antigen-specific T cell proliferation in vitro and lead to effective T cell stimulation and inhibition of tumor growth in vivo. FROM THE CLINICAL EDITOR Artifical antigen presenting cells could revolutionize the field of cancer-directed immunotherapy. This team of investigators have manufactured two types of nanoscale particle platform-based aAPCs and demonstrates that both iron-dextran particles and quantum dot nanocrystals enhance tumor rejection in a melanoma model, providing the first description of nanoscale aAPCs that lead to effective T cell stimulation and inhibition of tumor growth.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA; Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrés De León Medero
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Malarvizhi Durai
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yen Ling Chiu
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Joan Glick Bieler
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leah Sibener
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | - Michael Edidin
- Department of Biology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mathias Oelke
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jonathan Schneck
- Departments of Pathology, Oncology, and Medicine. Institute of Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
30
|
Sunshine JC, Green JJ. Nanoengineering approaches to the design of artificial antigen-presenting cells. Nanomedicine (Lond) 2013; 8:1173-89. [PMID: 23837856 PMCID: PMC3951141 DOI: 10.2217/nnm.13.98] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Artificial antigen-presenting cells (aAPCs) have shown great initial promise for ex vivo activation of cytotoxic T cells. The development of aAPCs has focused mainly on the choice of proteins to use for surface presentation to T cells when conjugated to various spherical, microscale particles. We review here biomimetic nanoengineering approaches that have been applied to the development of aAPCs that move beyond initial concepts about aAPC development. This article also discusses key technologies that may be enabling for the development of nano- and micro-scale aAPCs with nanoscale features, and suggests several future directions for the field.
Collapse
Affiliation(s)
- Joel C Sunshine
- Department of Biomedical Engineering & the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering & the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Wilmer Eye Institute & the Institute for Nanobiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
31
|
Perica K, Bieler JG, Edidin M, Schneck J. Modulation of MHC binding by lateral association of TCR and coreceptor. Biophys J 2013. [PMID: 23199917 DOI: 10.1016/j.bpj.2012.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The structure of a T cell receptor (TCR) and its affinity for cognate antigen are fixed, but T cells regulate binding sensitivity through changes in lateral membrane organization. TCR microclusters formed upon antigen engagement participate in downstream signaling. Microclusters are also found 3-4 days after activation, leading to enhanced antigen binding upon rechallenge. However, others have found an almost complete loss of antigen binding four days after T cell activation, when TCR clusters are present. To resolve these contradictory results, we compared binding of soluble MHC-Ig dimers by transgenic T cells stimulated with a high (100 μM) or low (100 fM) dose of cognate antigen. Cells activated by a high dose of peptide bound sixfold lower amounts of CD8-dependent ligand K(b)-SIY than cells activated by a low dose of MHC/peptide. In contrast, both cell populations bound a CD8-independent ligand L(d)-QL9 equally well. Consistent with the differences between binding of CD8-dependent and CD8-independent peptide/MHC, Förster resonance energy transfer (FRET) measurements of molecular proximity reported little nanoscale association of TCR with CD8 (16 FRET units) compared to their association on cells stimulated by low antigen dose (62 FRET units). Loss of binding induced by changes in lateral organization of TCR and CD8 may serve as a regulatory mechanism to avoid excessive inflammation and immunopathology in response to aggressive infection.
Collapse
Affiliation(s)
- Karlo Perica
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
32
|
Shen C, Cheng K, Miao S, Wang W, He Y, Meng F, Zhang J. Latex bead-based artificial antigen-presenting cells induce tumor-specific CTL responses in the native T-cell repertoires and inhibit tumor growth. Immunol Lett 2013; 150:1-11. [PMID: 23328744 DOI: 10.1016/j.imlet.2013.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 12/30/2012] [Accepted: 01/04/2013] [Indexed: 01/10/2023]
Abstract
Cell-free artificial antigen-presenting cells (aAPCs) were generated by coupling H-2K(b)/TRP2 tetramers together with anti-CD28 and anti-4-1BB antibodies onto cell-sized latex beads and injected intravenously and subcutaneously into naïve mice and antigen-primed mice (B6, H-2K(b)). Vigorous tumor antigen-specific CTL responses in the native T-cell repertoire in each mouse model were elicited as evaluated by measuring surface CD69 and CD25, intracellular IFN-γ, tetramer staining and cytolysis of melanoma cells. Furthermore, the aAPCs efficiently inhibited subcutaneous tumor growth and markedly delayed tumor progression in tumor-bearing mice. These data suggest that bead-based aAPCs represent a potential strategy for the active immunotherapy of cancers or persistent infections.
Collapse
Affiliation(s)
- Chuanlai Shen
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China.
| | | | | | | | | | | | | |
Collapse
|
33
|
Goldberg B, Bona C. Dimeric MHC-peptides inserted into an immunoglobulin scaffold as new immunotherapeutic agents. J Cell Mol Med 2011; 15:1822-32. [PMID: 21435177 PMCID: PMC3918039 DOI: 10.1111/j.1582-4934.2011.01319.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/17/2011] [Indexed: 11/28/2022] Open
Abstract
The interactions of the T cell receptor (TCR) with cognate MHC-peptide and co-stimulatory molecules expressed at surface of antigen presenting cells (APC) leads to activation or tolerance of T cells. The development of molecular biological tools allowed for the preparation of soluble MHC-peptide molecules as surrogate for the APC. A decade ago a monomeric class II MHC molecule in which the peptide was covalently linked to β-chain of class II molecule was generated. This type of molecule had a low-binding affinity and did not cause the multimerization of TCR. The requirement of multimerization of TCR led to development of a new class of reagents, chimeric peptides covalently linked to MHC that was dimerized via Fc fragment of an immunoglobulin and linked to 3' end of the β-chain of MHC class II molecule. These soluble dimerized MHC-peptide chimeric molecules display high affinity for the TCR and caused multimerization of TCR without processing by an APC. Because dimeric molecules are devoid of co-stimulatory molecules interacting with CD28, a second signal, they induce anergy rather the activation of T cells. In this review, we compare the human and murine dimerized MHC class II-peptides and their effect on CD4(+) T cells, particularly the generation of T regulatory cells, which make these chimeric molecules an appealing approach for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Burt Goldberg
- Department of Chemistry, New York University, NY 10003-6688, USA.
| | | |
Collapse
|
34
|
Lee JB, Oelke M, Ramachandra L, Canaday DH, Schneck JP. Decline of influenza-specific CD8+ T cell repertoire in healthy geriatric donors. IMMUNITY & AGEING 2011; 8:6. [PMID: 21846352 PMCID: PMC3179433 DOI: 10.1186/1742-4933-8-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 08/16/2011] [Indexed: 01/08/2023]
Abstract
Background While influenza vaccination results in protective antibodies against primary infections, clearance of infection is primarily mediated through CD8+ T cells. Studying the CD8+ T cell response to influenza epitopes is crucial in understanding the disease associated morbidity and mortality especially in at risk populations such as the elderly. We compared the CD8+ T cell response to immunodominant and subdominant influenza epitopes in HLA-A2+ control, adult donors, aged 21-42, and in geriatric donors, aged 65 and older. Results We used a novel artificial Antigen Presenting Cell (aAPC) based stimulation assay to reveal responses that could not be detected by enzyme-linked immunosorbent spot (ELISpot). 14 younger control donors and 12 geriatric donors were enrolled in this study. The mean number of influenza-specific subdominant epitopes per control donor detected by ELISpot was only 1.4 while the mean detected by aAPC assay was 3.3 (p = 0.0096). Using the aAPC assay, 92% of the control donors responded to at least one subdominant epitopes, while 71% of control donors responded to more than one subdominant influenza-specific response. 66% of geriatric donors lacked a subdominant influenza-specific response and 33% of geriatric donors responded to only 1 subdominant epitope. The difference in subdominant response between age groups is statistically significant (p = 0.0003). Conclusion Geriatric donors lacked the broad, multi-specific response to subdominant epitopes seen in the control donors. Thus, we conclude that aging leads to a decrease in the subdominant influenza-specific CTL responses which may contribute to the increased morbidity and mortality in older individuals.
Collapse
Affiliation(s)
- Jessica B Lee
- Department of Pathology, Johns Hopkins University, 733 N Broadway BRB 632, Baltimore, MD, 21205, USA.
| | | | | | | | | |
Collapse
|
35
|
Abstract
The observation that T cells can recognize and specifically eliminate cancer cells has spurred interest in the development of efficient methods to generate large numbers of T cells with specificity for tumor antigens that can be harnessed for use in cancer therapy. Recent studies have demonstrated that during encounter with tumor antigen, the signals delivered to T cells by professional antigen-presenting cells can affect T-cell programming and their subsequent therapeutic efficacy. This has stimulated efforts to develop artificial antigen-presenting cells that allow optimal control over the signals provided to T cells. In this review, we will discuss the advantages and disadvantages of cellular and acellular artificial antigen-presenting cell systems and their use in T-cell adoptive immunotherapy for cancer.
Collapse
|
36
|
Dynamic regulation of functionally distinct virus-specific T cells. Proc Natl Acad Sci U S A 2010; 107:3669-74. [PMID: 20133680 DOI: 10.1073/pnas.0915168107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The functional capacities of CD8(+) T cells important for virus clearance are influenced by interactions with antigen presenting cells (APCs) and CD4(+) T cells during initial selection, subsequent expansion, and development of memory. Recently, investigators have shown that polyfunctional T cells correlate best with long-term protection, however, it is still unknown how to stimulate T cells to achieve these responses. To study this, we examined the phenotypes and functions of CD8(+) T cells specific for two different virus antigens stimulated ex vivo using either autologous monocyte-derived dendritic cells (moDCs) or HLA-A2-Ig-based artificial APCs (aAPCs). Although similar numbers of influenza virus and measles virus tetramer-positive cells were generated by stimulation with peptide-loaded moDCs and aAPCs, T cell function, assessed by expression of IL-2, IFN-gamma, TNF-alpha, MIP1beta, and CD107a, showed that aAPC-generated CD8(+) T cells were multifunctional, whereas moDC-generated cells were mostly monofunctional. aAPC-generated cells also produced more of each cytokine per cell than CD8(+) T cells generated with moDCs. These phenotypes were not fixed, as changing the culture conditions of expanding T cells from aAPCs to moDCs, and moDCs to aAPCs, reversed the phenotypes. We conclude that CD8(+) T cells are heterogeneous in their functionality and that this is dependent, in a dynamic way, on the stimulating APC. These studies will lead to understanding the factors that influence induction of optimal CD8(+) T cell function.
Collapse
|
37
|
Steenblock ER, Wrzesinski SH, Flavell RA, Fahmy TM. Antigen presentation on artificial acellular substrates: modular systems for flexible, adaptable immunotherapy. Expert Opin Biol Ther 2010; 9:451-64. [PMID: 19344282 DOI: 10.1517/14712590902849216] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent findings on T cells and dendritic cells have elucidated principles that can be used for a bottom-up approach to engineering artificial antigen presentation on synthetic substrates. OBJECTIVE/METHODS To compare the latest artificial antigen-presenting cell (aAPC) technology, focussing on acellular systems because they offer advantages such as easy tunability and rapid point-of-care application compared with cellular systems. We review acellular aAPC performance and discuss their promise for clinical applications. RESULTS/CONCLUSION Acellular aAPCs are a powerful alternative to natural-cell-based therapies, offering flexibility and modularity for incorporation oSf a variety of stimuli, hence increasing precision. Current technologies should adapt physiologically important signals within safe materials to more closely approximate their cellular counterparts. These constructs could be administered parenterally as APC replacements for active vaccines or used ex vivo for adoptive immunotherapy.
Collapse
Affiliation(s)
- Erin R Steenblock
- Yale University, Malone Engineering Center, 55 Prospect Street, Room 402C, New Haven, CT 06511, USA
| | | | | | | |
Collapse
|
38
|
Webb TJ, Bieler JG, Schneck JP, Oelke M. Ex vivo induction and expansion of natural killer T cells by CD1d1-Ig coated artificial antigen presenting cells. J Immunol Methods 2009; 346:38-44. [PMID: 19446558 DOI: 10.1016/j.jim.2009.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Accepted: 05/04/2009] [Indexed: 10/20/2022]
Abstract
Natural killer T (NKT) cells play a pivotal role in maintaining immune homostasis. They recognize lipid antigen in the context of CD1d molecules and subsequently produce cytokines that activate cells of both the innate and adaptive immune responses. Many studies examining patients with autoimmune disease or cancer have shown that there is a reduction in both NKT cell number and function. Due to the complexities of manipulating NKT cells in vivo, ex vivo expanded effector NKT cells would be an excellent therapeutic modality. To date, immunotherapy utilizing the NKT/CD1d system has been dependent on the use of autologous DC in the presence or absence of a synthetic glycolipid, alpha-galactocylceramide. Here we report a novel technique that facilitates the growth and analysis of NKT cells through the use of CD1d-expressing aAPC. CD1d-based aAPC can effectively propagate both canonical (iNKT cells) and noncanonical (Valpha14(-)) NKT cells. Importantly, CD1d-Ig aAPC can expand NKT cells from cancer patients. Thus, CD1d-expressing aAPC will enhance our knowledge of NKT cell biology and could potentially be used as a novel tool in adoptive immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tonya J Webb
- Department of Pathology, Johns Hopkins School of Medicine, Ross Building Room 644S, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|