51
|
Engineering advanced dynamic biomaterials to optimize adoptive T-cell immunotherapy. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2021.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
52
|
Li W, Peng A, Wu H, Quan Y, Li Y, Lu L, Cui M. Anti-Cancer Nanomedicines: A Revolution of Tumor Immunotherapy. Front Immunol 2020; 11:601497. [PMID: 33408716 PMCID: PMC7779686 DOI: 10.3389/fimmu.2020.601497] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Immunotherapies have been accelerating the development of anti-cancer clinical treatment, but its low objective responses and severe off-target immune-related adverse events (irAEs) limit the range of application. Strategies to remove these obstacles primarily focus on the combination of different therapies and the exploitation of new immunotherapeutic agents. Nanomedicine potentiates the effects of activating immune cells selectively and reversing tumor induced immune deficiency microenvironment through multiple mechanisms. In the last decade, a variety of nano-enabled tumor immunotherapies was under clinical investigation. As time goes by, the advantages of nanomedicine are increasingly prominent. With the continuous development of nanotechnology, nanomedicine will offer more distinctive perspectives in imaging diagnosis and treatment of tumors. In this Review, we wish to provide an overview of tumor immunotherapy and the mechanisms of nanomaterials that aim to enhance the efficacy of tumor immunotherapy under development or in clinic treatment.
Collapse
Affiliation(s)
- Wei Li
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Anghui Peng
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Huajun Wu
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Yingyao Quan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Yong Li
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Min Cui
- Department of General Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| |
Collapse
|
53
|
Sui X, Jin T, Liu T, Wu S, Wu Y, Tang Z, Ren Y, Ni D, Yao Z, Zhang H. Tumor Immune Microenvironments (TIMEs): Responsive Nanoplatforms for Antitumor Immunotherapy. Front Chem 2020; 8:804. [PMID: 33094098 PMCID: PMC7508192 DOI: 10.3389/fchem.2020.00804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
Interest in cancer immunotherapy has rapidly risen since it offers many advantages over traditional approaches, such as high efficiency and prevention of metastasis. Efforts have primarily focused on two major strategies for regulating the body's antitumor immune response mechanisms: “enhanced immunotherapy” that aims to amplify the immune activation, and “normalized immunotherapy” that corrects the defective immune mechanism in the tumor immune microenvironments (TIMEs), which returns to the normal immune trajectory. However, due to the complexity and heterogeneity of the TIMEs, and lack of visualization research on the immunotherapy process, cancer immunotherapy has not been widely used in clinical setting. Recently, through the design and modification of nanomaterials, intelligent TIME-responsive nanoplatforms were developed from which encouraging results in many aspects of immunotherapy have been achieved. In this mini review, the status of designed nanomaterials for nanoplatform-based immune regulation of TIMEs has been emphasized, particularly with respect to the aforementioned approaches. It is envisaged that future prospects will focus on a combination of multiple immunotherapies for more efficient cancer inhibition and elimination.
Collapse
Affiliation(s)
- Xueqing Sui
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Teng Jin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tonghui Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiman Wu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yue Wu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongmin Tang
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Yan Ren
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dalong Ni
- Departments of Radiology, Medical Physics, and Pharmaceutical Sciences, University of Wisconsin - Madison, Madison, WI, United States
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua Zhang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
54
|
Abstract
Personalized cancer vaccines (PCVs) are reinvigorating vaccine strategies in cancer immunotherapy. In contrast to adoptive T-cell therapy and checkpoint blockade, the PCV strategy modulates the innate and adaptive immune systems with broader activation to redeploy antitumor immunity with individualized tumor-specific antigens (neoantigens). Following a sequential scheme of tumor biopsy, mutation analysis, and epitope prediction, the administration of neoantigens with synthetic long peptide (SLP) or mRNA formulations dramatically improves the population and activity of antigen-specific CD4+ and CD8+ T cells. Despite the promising prospect of PCVs, there is still great potential for optimizing prevaccination procedures and vaccine potency. In particular, the arduous development of tumor-associated antigen (TAA)-based vaccines provides valuable experience and rational principles for augmenting vaccine potency which is expected to advance PCV through the design of adjuvants, delivery systems, and immunosuppressive tumor microenvironment (TME) reversion since current personalized vaccination simply admixes antigens with adjuvants. Considering the broader application of TAA-based vaccine design, these two strategies complement each other and can lead to both personalized and universal therapeutic methods. Chemical strategies provide vast opportunities for (1) exploring novel adjuvants, including synthetic molecules and materials with optimizable activity, (2) constructing efficient and precise delivery systems to avoid systemic diffusion, improve biosafety, target secondary lymphoid organs, and enhance antigen presentation, and (3) combining bioengineering methods to innovate improvements in conventional vaccination, "smartly" re-educate the TME, and modulate antitumor immunity. As chemical strategies have proven versatility, reliability, and universality in the design of T cell- and B cell-based antitumor vaccines, the union of such numerous chemical methods in vaccine construction is expected to provide new vigor and vitality in cancer treatment.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.,Beijing Institute for Brain Disorders, 100069 Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, 100084 Beijing, China
| |
Collapse
|
55
|
Hickey JW, Isser A, Salathe SF, Gee KM, Hsiao MH, Shaikh W, Uzoukwu NC, Bieler JG, Mao HQ, Schneck JP. Adaptive Nanoparticle Platforms for High Throughput Expansion and Detection of Antigen-Specific T cells. NANO LETTERS 2020; 20:6289-6298. [PMID: 32594746 PMCID: PMC8008984 DOI: 10.1021/acs.nanolett.0c01511] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
T cells are critical players in disease; yet, their antigen-specificity has been difficult to identify, as current techniques are limited in terms of sensitivity, throughput, or ease of use. To address these challenges, we increased the throughput and translatability of magnetic nanoparticle-based artificial antigen presenting cells (aAPCs) to enrich and expand (E+E) murine or human antigen-specific T cells. We streamlined enrichment, expansion, and aAPC production processes by enriching CD8+ T cells directly from unpurified immune cells, increasing parallel processing capacity of aAPCs in a 96-well plate format, and designing an adaptive aAPC that enables multiplexed aAPC construction for E+E and detection. We applied these adaptive platforms to process and detect CD8+ T cells specific for rare cancer neoantigens, commensal bacterial cross-reactive epitopes, and human viral and melanoma antigens. These innovations dramatically increase the multiplexing ability and decrease the barrier to adopt for investigating antigen-specific T cell responses.
Collapse
Affiliation(s)
- John W. Hickey
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Ariel Isser
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sebastian F. Salathe
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Kayla M. Gee
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Meng-Hsuan Hsiao
- Department of Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Wasamah Shaikh
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nkechi C. Uzoukwu
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Joanie Glick Bieler
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hai-Quan Mao
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan P. Schneck
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
56
|
Sun L, Shen F, Xu J, Han X, Fan C, Liu Z. DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202003367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Jun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Shanghai Jiao Tong University Shanghai 201240 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices Soochow University Suzhou 215123 Jiangsu China
| |
Collapse
|
57
|
Acebes-Fernández V, Landeira-Viñuela A, Juanes-Velasco P, Hernández AP, Otazo-Perez A, Manzano-Román R, Gongora R, Fuentes M. Nanomedicine and Onco-Immunotherapy: From the Bench to Bedside to Biomarkers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1274. [PMID: 32610601 PMCID: PMC7407304 DOI: 10.3390/nano10071274] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
The broad relationship between the immune system and cancer is opening a new hallmark to explore for nanomedicine. Here, all the common and synergy points between both areas are reviewed and described, and the recent approaches which show the progress from the bench to the beside to biomarkers developed in nanomedicine and onco-immunotherapy.
Collapse
Affiliation(s)
- Vanessa Acebes-Fernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Angela-Patricia Hernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Andrea Otazo-Perez
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Raúl Manzano-Román
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| | - Rafael Gongora
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| |
Collapse
|
58
|
DNA‐Edited Ligand Positioning on Red Blood Cells to Enable Optimized T Cell Activation for Adoptive Immunotherapy. Angew Chem Int Ed Engl 2020; 59:14842-14853. [DOI: 10.1002/anie.202003367] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/25/2020] [Indexed: 12/21/2022]
|
59
|
Shi J, Clayton C, Tian B. Nano-enabled cellular engineering for bioelectric studies. NANO RESEARCH 2020; 13:1214-1227. [PMID: 34295455 PMCID: PMC8294124 DOI: 10.1007/s12274-019-2580-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/24/2019] [Indexed: 06/13/2023]
Abstract
Engineered cells have opened up a new avenue for scientists and engineers to achieve specialized biological functions. Nanomaterials, such as silicon nanowires and quantum dots, can establish tight interfaces with cells either extra- or intracellularly, and they have already been widely used to control cellular functions. The future exploration of nanomaterials in cellular engineering may reveal numerous opportunities in both fundamental bioelectric studies and clinic applications. In this review, we highlight several nanomaterials-enabled non-genetic approaches to fabricating engineered cells. First, we briefly review the latest progress in engineered or synthetic cells, such as protocells that create cell-like behaviors from nonliving building blocks, and cells made by genetic or chemical modifications. Next, we illustrate the need for non-genetic cellular engineering with semiconductors and present some examples where chemical synthesis yields complex morphology or functions needed for biointerfaces. We then provide discussions in detail about the semiconductor nanostructure-enabled neural, cardiac, and microbial modulations. We also suggest the need to integrate tissue engineering with semiconductor devices to carry out more complex functions. We end this review by providing our perspectives for future development in non-genetic cellular engineering.
Collapse
Affiliation(s)
- Jiuyun Shi
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | | | - Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
60
|
Huang Y, Zeng J. Recent development and applications of nanomaterials for cancer
immunotherapy. NANOTECHNOLOGY REVIEWS 2020; 9:367-384. [DOI: 10.1515/ntrev-2020-0027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Immunotherapy, which utilizes the patient’s own immune system to fight against
cancer, further results in durable antitumor responses and reduces metastasis and
recurrence, has become one of the most effective and important cancer therapies along
with surgery, radiotherapy, and chemotherapy. Nanomaterials with the advantages of
large specific surface, delivery function, and controllable surface chemistry are
used to deliver antigens or adjuvants, or both, help to boost immune responses with
the imaging function or just act as adjuvants themselves and modulate tumor
microenvironment (TME). In this review, recent development and applications of
nanomaterials for cancer immunotherapy including delivery systems based on
nanomaterials, uniting imaging, self-adjuvants, targeting functions, artificial
antigen presenting cells, and TME modulation are focused and discussed.
Collapse
Affiliation(s)
- Yao Huang
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| | - Jinhua Zeng
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005 , China
| |
Collapse
|
61
|
Bessell CA, Isser A, Havel JJ, Lee S, Bell DR, Hickey JW, Chaisawangwong W, Glick Bieler J, Srivastava R, Kuo F, Purohit T, Zhou R, Chan TA, Schneck JP. Commensal bacteria stimulate antitumor responses via T cell cross-reactivity. JCI Insight 2020; 5:135597. [PMID: 32324171 DOI: 10.1172/jci.insight.135597] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/25/2020] [Indexed: 12/15/2022] Open
Abstract
Recent studies show gut microbiota modulate antitumor immune responses; one proposed mechanism is cross-reactivity between antigens expressed in commensal bacteria and neoepitopes. We found that T cells targeting an epitope called SVYRYYGL (SVY), expressed in the commensal bacterium Bifidobacterium breve (B. breve), cross-react with a model neoantigen, SIYRYYGL (SIY). Mice lacking B. breve had decreased SVY-reactive T cells compared with B. breve-colonized mice, and the T cell response was transferable by SVY immunization or by cohousing mice without Bifidobacterium with ones colonized with Bifidobacterium. Tumors expressing the model SIY neoantigen also grew faster in mice lacking B. breve compared with Bifidobacterium-colonized animals. B. breve colonization also shaped the SVY-reactive TCR repertoire. Finally, SVY-specific T cells recognized SIY-expressing melanomas in vivo and led to decreased tumor growth and extended survival. Our work demonstrates that commensal bacteria can stimulate antitumor immune responses via cross-reactivity and how bacterial antigens affect the T cell landscape.
Collapse
Affiliation(s)
| | - Ariel Isser
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan J Havel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sangyun Lee
- Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, New York, USA
| | - David R Bell
- Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, New York, USA
| | - John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Worarat Chaisawangwong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joan Glick Bieler
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raghvendra Srivastava
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Fengshen Kuo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tanaya Purohit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ruhong Zhou
- Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, New York, USA.,Department of Chemistry, Columbia University, New York, New York, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jonathan P Schneck
- Graduate Program in Immunology and.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Institute of Cellular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
62
|
Ichikawa J, Yoshida T, Isser A, Laino AS, Vassallo M, Woods D, Kim S, Oelke M, Jones K, Schneck JP, Weber JS. Rapid Expansion of Highly Functional Antigen-Specific T Cells from Patients with Melanoma by Nanoscale Artificial Antigen-Presenting Cells. Clin Cancer Res 2020; 26:3384-3396. [PMID: 32241816 DOI: 10.1158/1078-0432.ccr-19-3487] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/13/2020] [Accepted: 03/30/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE Generation of antigen-specific T cells from patients with cancer employs large numbers of peripheral blood cells and/or tumor-infiltrating cells to generate antigen-presenting and effector cells commonly requiring multiple rounds of restimulation ex vivo. We used a novel paramagnetic, nanoparticle-based artificial antigen-presenting cell (nano-aAPC) that combines anti-CD28 costimulatory and human MHC class I molecules that are loaded with antigenic peptides to rapidly expand tumor antigen-specific T cells from patients with melanoma. EXPERIMENTAL DESIGN Nano-aAPC-expressing HLA-A*0201 molecules and costimulatory anti-CD28 antibody and HLA-A*0201 molecules loaded with MART-1 or gp100 class I-restricted peptides were used to stimulate CD8 T cells purified from the peripheral blood of treatment-naïve or PD-1 antibody-treated patients with stage IV melanoma. Expanded cells were restimulated with fresh peptide-pulsed nano-aAPC at day 7. Phenotype analysis and functional assays including cytokine release, cytolysis, and measurement of avidity were conducted. RESULTS MART-1-specific CD8 T cells rapidly expanded up to 1,000-fold by day 14 after exposure to peptide-pulsed nano-aAPC. Expanded T cells had a predominantly stem cell memory CD45RA+/CD62L+/CD95+ phenotype; expressed ICOS, PD-1, Tim3, and LAG3; and lacked CD28. Cells from patients with melanoma were polyfunctional; highly avid; expressed IL2, IFNγ, and TNFα; and exhibited cytolytic activity against tumor cell lines. They expanded 2- to 3-fold after exposure to PD-1 antibody in vivo, and expressed a highly diverse T-cell receptor V beta repertoire. CONCLUSIONS Peptide-pulsed nano-aAPC rapidly expanded polyfunctional antigen-specific CD8 T cells with high avidity, potent lytic function, and a stem cell memory phenotype from patients with melanoma.
Collapse
Affiliation(s)
- Junya Ichikawa
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York.
| | - Tatsuya Yoshida
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - Ariel Isser
- Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Andressa S Laino
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - Melinda Vassallo
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | - David Woods
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York
| | | | | | | | | | - Jeffrey S Weber
- NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, New York, New York.
| |
Collapse
|
63
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
64
|
Biomaterials for Immunoengineering. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00076-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
65
|
Mukherjee S, Sonanini D, Maurer A, Daldrup-Link HE. The yin and yang of imaging tumor associated macrophages with PET and MRI. Am J Cancer Res 2019; 9:7730-7748. [PMID: 31695797 PMCID: PMC6831464 DOI: 10.7150/thno.37306] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor associated macrophages (TAM) are key players in the cancer microenvironment. Molecular imaging modalities such as MRI and PET can be used to track and monitor TAM dynamics in tumors non-invasively, based on specific uptake and quantification of MRI-detectable nanoparticles or PET-detectable radiotracers. Particular molecular signatures can be leveraged to target anti-inflammatory TAM, which support tumor growth, and pro-inflammatory TAM, which suppress tumor growth. In addition, TAM-directed imaging probes can be designed to include immune modulating properties, thereby leading to combined diagnostic and therapeutic (theranostic) effects. In this review, we will discuss the complementary role of TAM-directed radiotracers and iron oxide nanoparticles for monitoring cancer immunotherapies with PET and MRI technologies. In addition, we will outline how TAM-directed imaging and therapy is interdependent and can be connected towards improved clinical outcomes
Collapse
|
66
|
Li Y, Ayala-Orozco C, Rauta PR, Krishnan S. The application of nanotechnology in enhancing immunotherapy for cancer treatment: current effects and perspective. NANOSCALE 2019; 11:17157-17178. [PMID: 31531445 PMCID: PMC6778734 DOI: 10.1039/c9nr05371a] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer immunotherapy is emerging as a promising treatment modality that suppresses and eliminates tumors by re-activating and maintaining the tumor-immune cycle, and further enhancing the body's anti-tumor immune response. Despite the impressive therapeutic potential of immunotherapy approaches such as immune checkpoint inhibitors and tumor vaccines in pre-clinical and clinical applications, the effective response is limited by insufficient accumulation in tumor tissues and severe side-effects. Recent years have witnessed the rise of nanotechnology as a solution to improve these technical weaknesses due to its inherent biophysical properties and multifunctional modifying potential. In this review, we summarized and discussed the current status of nanoparticle-enhanced cancer immunotherapy strategies, including intensified delivery of tumor vaccines and immune adjuvants, immune checkpoint inhibitor vehicles, targeting capacity to tumor-draining lymph nodes and immune cells, triggered releasing and regulating specific tumor microenvironments, and adoptive cell therapy enhancement effects.
Collapse
Affiliation(s)
- Yongjiang Li
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China. and Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Ciceron Ayala-Orozco
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Pradipta Ranjan Rauta
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Sunil Krishnan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA. and Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
67
|
Abstract
The 2018 Nobel Prize in Physiology or Medicine was awarded to pioneers in the field of cancer immunotherapy, as the utility of leveraging a patient's coordinated and adaptive immune system to fight the patient's unique tumour has now been validated robustly in the clinic. Still, the proportion of patients who respond to immunotherapy remains modest (~15% objective response rate across indications), as tumours have multiple means of immune evasion. The immune system is spatiotemporally controlled, so therapies that influence the immune system should be spatiotemporally controlled as well, in order to maximize the therapeutic index. Nanoparticles and biomaterials enable one to program the location, pharmacokinetics and co-delivery of immunomodulatory compounds, eliciting responses that cannot be achieved upon administration of such compounds in solution. The convergence of cancer immunotherapy, nanotechnology, bioengineering and drug delivery is opportune, as each of these fields has matured independently to the point that it can now be used to complement the others substantively and rationally, rather than modestly and empirically. As a result, unmet needs increasingly can be addressed with deductive intention. This Review explores how nanotechnology and related approaches are being applied to augmenting both endogenous leukocytes and adoptively transferred ones by informing specificity, influencing localization and improving function.
Collapse
|
68
|
McBride DA, Kerr MD, Wai SL, Shah NJ. Applications of molecular engineering in T-cell-based immunotherapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1557. [PMID: 30972976 PMCID: PMC7869905 DOI: 10.1002/wnan.1557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/24/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023]
Abstract
Harnessing an individual's immune cells to mediate antitumor and antiviral responses is a life-saving option for some patients with otherwise intractable forms of cancer and infectious disease. In particular, T-cell-based engineered immune cells are a powerful new class of therapeutics with remarkable efficacy. Clinical experience has helped to define some of the major challenges for reliable, safe, and effective deployment of T-cells against a broad range of diseases. While poised to revolutionize immunotherapy, scalable manufacturing, safety, specificity, and the development of resistance are potential roadblocks in their widespread usage. The development of molecular engineering tools to allow for the direct or indirect engineering of T-cells to enable one to troubleshoot delivery issues, amplify immunomodulatory effects, integrate the synergistic effects of different molecules, and home to the target cells in vivo. In this review, we will analyze thus-far developed cell- and material-based tools for enhancing T-cell therapies, including methods to improve safety and specificity, enhancing efficacy, and overcoming limitations in scalable manufacturing. We summarize the potential of T-cells as immune modulating therapies and the potential future directions for enabling their adoption for a broad range of diseases. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Cells at the Nanoscale.
Collapse
Affiliation(s)
- David A McBride
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Matthew D Kerr
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Shinya L Wai
- Department of Nanoengineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
| | - Nisarg J Shah
- Department of Nanoengineering, University of California, San Diego, California
- Program in Chemical Engineering, University of California, San Diego, California
- Center for Nano-Immuno Engineering, University of California, San Diego, California
- Graduate Program in Immunology, University of California, San Diego, California
- San Diego Center for Precision Immunotherapy, University of California, San Diego, California
| |
Collapse
|
69
|
Bai Y, Wang Y, Zhang X, Fu J, Xing X, Wang C, Gao L, Liu Y, Shi L. Potential applications of nanoparticles for tumor microenvironment remodeling to ameliorate cancer immunotherapy. Int J Pharm 2019; 570:118636. [PMID: 31446027 DOI: 10.1016/j.ijpharm.2019.118636] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
In recent years, researchers have made significant innovations in the field of tumor immunotherapy based on the knowledge of biology, oncology, and immunology. Tumor immunotherapy involves the use of immune checkpoint inhibitors and CAR (chimeric antigen receptor)-T cell therapy. As compared with conventional chemotherapy, immunotherapy is a potential approach to induce a more powerful immune response against tumor in the patient suffering from the advanced stage malignancy. Regardless of the developments made, a large number of clinical studies have confirmed that a substantial number of cancer patients still demonstrate non-responsiveness to immunotherapy, mainly due to the immunomodulating interactions of tumor cells with the immunosuppressive tumor microenvironment (iTME). It leads to immune tolerance of tumors and influences the efficacy of immunotherapy. This immune failure could be attributed to a complex immunosuppressive network comprising stromal and inflammatory cells, vessel system, ECM (extracellular matrix) and the cytokines released in tumor microenvironment (TME). The antitumor immune activity can be enhanced at different stages of tumor development by selective suppression of inhibitory pathways in the TME. This specific task can be achieved by using nano-sized drug delivery tools which are specific in their action and biocompatible in nature. Several recent studies have described the use of nanoparticles for iTME remodeling through the specific elimination of immunosuppressive cells, obstructing immune checkpoints, promotion of inflammatory cytokines, and amending the regulatory cells of the immune system. The efficacy of current immunotherapy can be improved by nanoparticle-mediated remodeling of iTME.
Collapse
Affiliation(s)
- Yuzhuo Bai
- Extrathoracic and Thyroid Mammary Surgery, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yun Wang
- Extrathoracic and Thyroid Mammary Surgery, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xudong Zhang
- Encephalopathy Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jianhua Fu
- Reproductive Center, Jilin Provincial People's Hospital, Changchun 130021, China
| | - Xiuli Xing
- Physical Examination Center, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Chunlan Wang
- Respiratory Department, First Clinical College, Academy of Traditional Chinese Medicine, Changchun 130021, China
| | - Longlan Gao
- Brain Surgery, Liaoyuan Hospital of Traditional Chinese Medicine, Liaoyuan 136000, China
| | - Yu Liu
- Brain Surgery, Liaoyuan Hospital of Traditional Chinese Medicine, Liaoyuan 136000, China
| | - Li Shi
- Department of Oncology and Hematology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China.
| |
Collapse
|
70
|
Liu J, Zhang R, Xu ZP. Nanoparticle-Based Nanomedicines to Promote Cancer Immunotherapy: Recent Advances and Future Directions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900262. [PMID: 30908864 DOI: 10.1002/smll.201900262] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/21/2019] [Indexed: 05/27/2023]
Abstract
Cancer immunotherapy is a promising cancer terminator by directing the patient's own immune system in the fight against this challenging disorder. Despite the monumental therapeutic potential of several immunotherapy strategies in clinical applications, the efficacious responses of a wide range of immunotherapeutic agents are limited in virtue of their inadequate accumulation in the tumor tissue and fatal side effects. In the last decades, increasing evidences disclose that nanotechnology acts as an appealing solution to address these technical barriers via conferring rational physicochemical properties to nanomaterials. In this Review, an imperative emphasis will be drawn from the current understanding of the effect of a nanosystem's structure characteristics (e.g., size, shape, surface charge, elasticity) and its chemical modification on its transport and biodistribution behavior. Subsequently, rapid-moving advances of nanoparticle-based cancer immunotherapies are summarized from traditional vaccine strategies to recent novel approaches, including delivery of immunotherapeutics (such as whole cancer cell vaccines, immune checkpoint blockade, and immunogenic cell death) and engineered immune cells, to regulate tumor microenvironment and activate cellular immunity. The future prospects may involve in the rational combination of a few immunotherapies for more efficient cancer inhibition and elimination.
Collapse
Affiliation(s)
- Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
71
|
Phuengkham H, Ren L, Shin IW, Lim YT. Nanoengineered Immune Niches for Reprogramming the Immunosuppressive Tumor Microenvironment and Enhancing Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803322. [PMID: 30773696 DOI: 10.1002/adma.201803322] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 01/03/2019] [Indexed: 06/09/2023]
Abstract
Cancer immunotherapies that harness the body's immune system to combat tumors have received extensive attention and become mainstream strategies for treating cancer. Despite promising results, some problems remain, such as the limited patient response rate and the emergence of severe immune-related adverse effects. For most patients, the therapeutic efficacy of cancer immunotherapy is mainly limited by the immunosuppressive tumor microenvironment (TME). To overcome such obstacles in the TME, the immunomodulation of immunosuppressive factors and therapeutic immune cells (e.g., T cells and antigen-presenting cells) should be carefully designed and evaluated. Nanoengineered synthetic immune niches have emerged as highly customizable platforms with a potent capability for reprogramming the immunosuppressive TME. Here, recent developments in nano-biomaterials that are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy which are rationally designed to modulate the immunosuppressive TME in a spatiotemporal manner for enhanced cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Hathaichanok Phuengkham
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Long Ren
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Il Woo Shin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| |
Collapse
|
72
|
Chiozzini C, Olivetta E, Sanchez M, Arenaccio C, Ferrantelli F, Leone P, Federico M. Tumor cells endowed with professional antigen-presenting cell functions prime PBLs to generate antitumor CTLs. J Mol Med (Berl) 2019; 97:1139-1153. [PMID: 31161312 DOI: 10.1007/s00109-019-01797-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Intrinsic genetic instability of tumor cells leads to continuous production of mutated proteins referred to as tumor-specific neoantigens. Generally, they are recognized as nonself products by the host immune system. However, an effective adaptive response clearing neoantigen-expressing cells is lost in tumor diseases. Most advanced therapeutic strategies aim at inducing neoantigen-specific immune activation through personalized approaches. They include tumor cell exome sequencing, human leukocyte antigen (HLA) typing, synthesis, and injection of peptides/RNA with adjuvants. Here, we propose an innovative method to induce a CD8+ T cytotoxic lymphocyte (CTL) immune response against tumor neoantigens bypassing the steps needed in current therapeutic strategies of personalized vaccination. We assumed that tumor cells can be the most efficient and precise factory of major histocompatibility complex (MHC) class I-associated, tumor neoantigen-derived peptides. Hence, endowing tumor cells with professional antigen-presenting functions would prime CD8+ T lymphocytes towards a response against nonself tumor antigens. To explore this possibility, both adenocarcinoma and melanoma human cells were engineered to express both CD80 and CD86 costimulatory molecules. HLA-matched lymphocytes were then primed through cocultivation with the engineered tumor cells. The generation of tumor-specific CD8+ T lymphocytes was tested through the combined analysis of cell activation markers, formation of immunologic synapses, generation of tumor antigen-specific CD8+ T lymphocytes, and cytotoxic activity. Our data consistently indicate that tumor cells endowed with professional antigen-presenting functions can generate an effective tumor-specific CTL immune response. This finding may open avenues towards the development of innovative antitumor immunotherapies. KEY MESSAGES: We established a novel method to induce antitumor CTLs without a need to identify TAAs and/or tumor neoantigens. This strategy relies on transducing tumor cells with a retroviral vector expressing both CD80 and CD86. In this way, tumor cells prime naïve CD8+ T lymphocytes in a way that CTLs killing the same tumor cells are generated. These findings open the way towards preclinical assays in the perspective to introduce this antitumor immunotherapy strategy in clinic.
Collapse
Affiliation(s)
- Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Eleonora Olivetta
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Massimo Sanchez
- Core Facilities, ISS, Viale Regina Elena 299, 00161, Rome, Italy
| | - Claudia Arenaccio
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Flavia Ferrantelli
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Patrizia Leone
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
73
|
Engineered biomaterials to mitigate growth factor cost in cell biomanufacturing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
74
|
Hickey JW, Dong Y, Chung JW, Salathe SF, Pruitt HC, Li X, Chang C, Fraser AK, Bessell CA, Ewald AJ, Gerecht S, Mao HQ, Schneck JP. Engineering an Artificial T-Cell Stimulating Matrix for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807359. [PMID: 30968468 PMCID: PMC8601018 DOI: 10.1002/adma.201807359] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/04/2019] [Indexed: 05/17/2023]
Abstract
T cell therapies require the removal and culture of T cells ex vivo to expand several thousand-fold. However, these cells often lose the phenotype and cytotoxic functionality for mediating effective therapeutic responses. The extracellular matrix (ECM) has been used to preserve and augment cell phenotype; however, it has not been applied to cellular immunotherapies. Here, a hyaluronic acid (HA)-based hydrogel is engineered to present the two stimulatory signals required for T-cell activation-termed an artificial T-cell stimulating matrix (aTM). It is found that biophysical properties of the aTM-stimulatory ligand density, stiffness, and ECM proteins-potentiate T cell signaling and skew phenotype of both murine and human T cells. Importantly, the combination of the ECM environment and mechanically sensitive TCR signaling from the aTM results in a rapid and robust expansion of rare, antigen-specific CD8+ T cells. Adoptive transfer of these tumor-specific cells significantly suppresses tumor growth and improves animal survival compared with T cells stimulated by traditional methods. Beyond immediate immunotherapeutic applications, demonstrating the environment influences the cellular therapeutic product delineates the importance of the ECM and provides a case study of how to engineer ECM-mimetic materials for therapeutic immune stimulation in the future.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Institute for Cell Engineering, School of Medicine, Baltimore, MD, 21205, USA
- Department of Pathology, School of Medicine, Baltimore, MD, 21287, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
| | - Yi Dong
- Graduate Program in Immunology, School of Medicine, Baltimore, MD, 21205, USA
| | - Jae Wook Chung
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Sebastian F Salathe
- Department of Biology, Krieger School of Arts and Sciences, Baltimore, MD, 21218, USA
| | - Hawley C Pruitt
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Xiaowei Li
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Calvin Chang
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
| | - Andrew K Fraser
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Baltimore, MD, 21205, USA
| | - Catherine A Bessell
- Graduate Program in Immunology, School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew J Ewald
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Baltimore, MD, 21205, USA
- Department of Oncology, School of Medicine, Baltimore, MD, 21205, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
- Physical Sciences-Oncology Center, Baltimore, MD, 21218, USA
| | - Hai-Quan Mao
- Department of Biomedical Engineering, School of Medicine, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Baltimore, MD, 21218, USA
| | - Jonathan P Schneck
- Institute for Cell Engineering, School of Medicine, Baltimore, MD, 21205, USA
- Department of Pathology, School of Medicine, Baltimore, MD, 21287, USA
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| |
Collapse
|
75
|
Kim S, Shah SB, Graney PL, Singh A. Multiscale engineering of immune cells and lymphoid organs. NATURE REVIEWS. MATERIALS 2019; 4:355-378. [PMID: 31903226 PMCID: PMC6941786 DOI: 10.1038/s41578-019-0100-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Immunoengineering applies quantitative and materials-based approaches for the investigation of the immune system and for the development of therapeutic solutions for various diseases, such as infection, cancer, inflammatory diseases and age-related malfunctions. The design of immunomodulatory and cell therapies requires the precise understanding of immune cell formation and activation in primary, secondary and ectopic tertiary immune organs. However, the study of the immune system has long been limited to in vivo approaches, which often do not allow multidimensional control of intracellular and extracellular processes, and to 2D in vitro models, which lack physiological relevance. 3D models built with synthetic and natural materials enable the structural and functional recreation of immune tissues. These models are being explored for the investigation of immune function and dysfunction at the cell, tissue and organ levels. In this Review, we discuss 2D and 3D approaches for the engineering of primary, secondary and tertiary immune structures at multiple scales. We highlight important insights gained using these models and examine multiscale engineering strategies for the design and development of immunotherapies. Finally, dynamic 4D materials are investigated for their potential to provide stimuli-dependent and context-dependent scaffolds for the generation of immune organ models.
Collapse
Affiliation(s)
- Sungwoong Kim
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Shivem B. Shah
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Pamela L. Graney
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- These authors contributed equally: Sungwoong Kim, Shivem B. Shah, Pamela L. Graney
| | - Ankur Singh
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
76
|
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]
|
77
|
Mi Y, Hagan CT, Vincent BG, Wang AZ. Emerging Nano-/Microapproaches for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801847. [PMID: 30937265 PMCID: PMC6425500 DOI: 10.1002/advs.201801847] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/01/2018] [Indexed: 05/15/2023]
Abstract
Cancer immunotherapy has achieved remarkable clinical efficacy through recent advances such as chimeric antigen receptor-T cell (CAR-T) therapy, immune checkpoint blockade (ICB) therapy, and neoantigen vaccines. However, application of immunotherapy in a clinical setting has been limited by low durable response rates and immune-related adverse events. The rapid development of nano-/microtechnologies in the past decade provides potential strategies to improve cancer immunotherapy. Advances of nano-/microparticles such as virus-like size, high surface to volume ratio, and modifiable surfaces for precise targeting of specific cell types can be exploited in the design of cancer vaccines and delivery of immunomodulators. Here, the emerging nano-/microapproaches in the field of cancer vaccines, immune checkpoint blockade, and adoptive or indirect immunotherapies are summarized. How nano-/microparticles improve the efficacy of these therapies, relevant immunological mechanisms, and how nano-/microparticle methods are able to accelerate the clinical translation of cancer immunotherapy are explored.
Collapse
Affiliation(s)
- Yu Mi
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - C. Tilden Hagan
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer CenterDepartment of Microbiology & ImmunologyCurriculum in Bioinformatics and Computational BiologyDivision of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Andrew Z. Wang
- Laboratory of Nano‐ and Translational MedicineCarolina Center for Cancer Nanotechnology ExcellenceCarolina Institute of NanomedicineLineberger Comprehensive Cancer CenterDepartment of Radiation OncologyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| |
Collapse
|
78
|
Huang P, Wang X, Liang X, Yang J, Zhang C, Kong D, Wang W. Nano-, micro-, and macroscale drug delivery systems for cancer immunotherapy. Acta Biomater 2019; 85:1-26. [PMID: 30579043 DOI: 10.1016/j.actbio.2018.12.028] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022]
Abstract
Immunotherapy is moving to the frontier of cancer treatment. Drug delivery systems (DDSs) have greatly advanced the development of cancer immunotherapeutic regimen and combination treatment. DDSs can spatiotemporally present tumor antigens, drugs, immunostimulatory molecules, or adjuvants, thus enabling the modulation of immune cells including dendritic cells (DCs) or T-cells directly in vivo and thereby provoking robust antitumor immune responses. Cancer vaccines, immune checkpoint blockade, and adoptive cell transfer have shown promising therapeutic efficiency in clinic, and the incorporation of DDSs may further increase antitumor efficiency while decreasing adverse side effects. This review focuses on the use of nano-, micro-, and macroscale DDSs for co-delivery of different immunostimulatory factors to reprogram the immune system to combat cancer. Regarding to nanoparticle-based DDSs, we emphasize the nanoparticle-based tumor immune environment modulation or as an addition to gene therapy, photodynamic therapy, or photothermal therapy. For microparticle or capsule-based DDSs, an overview of the carrier type, fabrication approach, and co-delivery of tumor vaccines and adjuvants is introduced. Finally, macroscale DDSs including hydrogels and scaffolds are also included and their role in personalized vaccine delivery and adoptive cell transfer therapy are described. Perspective and clinical translation of DDS-based cancer immunotherapy is also discussed. We believe that DDSs hold great potential in advancing the fundamental research and clinical translation of cancer immunotherapy. STATEMENT OF SIGNIFICANCE: Immunotherapy is moving to the frontier of cancer treatment. Drug delivery systems (DDSs) have greatly advanced the development of cancer immunotherapeutic regimen and combination treatment. In this comprehensive review, we focus on the use of nano-, micro-, and macroscale DDSs for the co-delivery of different immunostimulatory factors to reprogram the immune system to combat cancer. We also propose the perspective on the development of next-generation DDS-based cancer immunotherapy. This review indicates that DDSs can augment the antitumor T-cell immunity and hold great potential in advancing the fundamental research and clinical translation of cancer immunotherapy by simultaneously delivering dual or multiple immunostimulatory drugs.
Collapse
|
79
|
Sang W, Zhang Z, Dai Y, Chen X. Recent advances in nanomaterial-based synergistic combination cancer immunotherapy. Chem Soc Rev 2019; 48:3771-3810. [DOI: 10.1039/c8cs00896e] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This review aims to summarize various synergistic combination cancer immunotherapy strategies based on nanomaterials.
Collapse
Affiliation(s)
- Wei Sang
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Zhan Zhang
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Yunlu Dai
- Cancer Centre
- Faculty of Health Sciences
- University of Macau
- Macau SAR 999078
- China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine
- National Institute of Biomedical Imaging and Bioengineering
- National Institutes of Health
- Bethesda
- USA
| |
Collapse
|
80
|
Gao S, Yang D, Fang Y, Lin X, Jin X, Wang Q, Wang X, Ke L, Shi K. Engineering Nanoparticles for Targeted Remodeling of the Tumor Microenvironment to Improve Cancer Immunotherapy. Theranostics 2019; 9:126-151. [PMID: 30662558 PMCID: PMC6332787 DOI: 10.7150/thno.29431] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/01/2018] [Indexed: 12/22/2022] Open
Abstract
Owing to the fast-paced growth and cross-infiltration of oncology, immunology and molecular biology, tumor immunotherapy technology represented by immune checkpoint blockade and chimeric antigen receptor (CAR) T cell therapy has lately made remarkable advancements. In comparison with traditional chemotherapy, immunotherapy has the potential to elicit a stronger sustained antitumor immune response in those patients who have advanced malignant malignancies. In spite of the advancements made, a significant number of clinical research works have validated that an extensive proportion of cancer patients still manifest insensitivity to immunotherapy, primarily because of the immunomodulatory interactions between tumor cells and the immunosuppressive tumor microenvironment (TME), together mediating the immune tolerance of tumors and accordingly impacting the positive response to immunotherapy. The intricate immunosuppressive networks formed by stromal cells, inflammatory cells, vasculature, extracellular matrix (ECM), and their secreted cytokines in the TME, play a pivotal role in tumor immune escape. Specific blocking of inhibition pathways in the TME is expected to effectively prevent immune escape and tolerance of tumor cells in addition to their metastasis, accordingly improving the antitumor immune response at various phases of tumor growth. Emerging nanoscale targeted drug carriers truly suit this specific requirement due to their specificity, biocompatibility, and convenience of production. This review emphasizes recent attempts to remodel the tumor immune microenvironment using novel nanoparticles, which include specifically eliminating immunosuppressive cells, reprogramming immune regulatory cells, promoting inflammatory cytokines and blocking immune checkpoints. Targeted remodeling of the immunosuppressive TME using well-designed and fabricated nanoparticles provides a promising strategy for improving the effectiveness of current immunotherapy and is greatly significant.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Dongjuan Yang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Yan Fang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xiaojie Lin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xuechao Jin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Qi Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Liyuan Ke
- Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, P. R. China
| | - Kai Shi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| |
Collapse
|
81
|
Isser A, Schneck JP. High-affinity T cell receptors for adoptive cell transfer. J Clin Invest 2018; 129:69-71. [PMID: 30530992 DOI: 10.1172/jci125471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adoptive cell transfer (ACT) of engineered T cell receptors (TCRs) for cancer immunotherapy has evolved from simple gene transfer of isolated TCRs to various affinity enhancement techniques that overcome limitations imposed by central and peripheral tolerance on TCR affinity. In the current issue of the JCI, Poncette et al. used mice with human TCRαβ and HLA gene loci to discover CD4+ TCRs of optimal affinity for cancer testis antigen (CTA) NY-ESO-1. They combined this TCR with a previously discovered NY-ESO-1-specific CD8+ TCR in an ACT fibrosarcoma tumor model to demonstrate the importance of T cell help in mediating antitumor responses.
Collapse
Affiliation(s)
| | - Jonathan P Schneck
- Department of Pathology, Medicine and Oncology, and.,Immunology Program, Institute of Cellular Engineering, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
82
|
Hickey JW, Schneck JP. Enrich and Expand Rare Antigen-specific T Cells with Magnetic Nanoparticles. J Vis Exp 2018. [PMID: 30507913 DOI: 10.3791/58640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We have developed a tool to both enrich and expand antigen-specific T cells. This can be helpful in cases such as to A) detect the existence of antigen-specific T cells, B) probe the dynamics of antigen-specific responses, C) understand how antigen-specific responses affect disease state such as autoimmunity, D) demystify heterogeneous responses for antigen-specific T cells, or E) utilize antigen-specific cells for therapy. The tool is based on a magnetic particle that we conjugate antigen-specific and T cell co-stimulatory signals, and that we term as artificial antigen presenting cells (aAPCs). Consequently, since the technology is simple to produce, it can easily be adopted by other laboratories; thus, our purpose here is to describe in detail the fabrication and subsequent use of the aAPCs. We explain how to attach antigen-specific and co-stimulatory signals to the aAPCs, how to utilize them to enrich for antigen-specific T cells, and how to expand antigen-specific T cells. Furthermore, we will highlight engineering design considerations based on experimental and biological information of our experience with characterizing antigen-specific T cells.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University; Institute for Cell Engineering, School of Medicine, Johns Hopkins University; Institute for Nanobiotechnology, Johns Hopkins University; Department of Pathology, School of Medicine, Johns Hopkins University
| | - Jonathan P Schneck
- Institute for Cell Engineering, School of Medicine, Johns Hopkins University; Department of Pathology, School of Medicine, Johns Hopkins University;
| |
Collapse
|
83
|
Liu Z, Chen X, Zhang Z, Zhang X, Saunders L, Zhou Y, Ma PX. Nanofibrous Spongy Microspheres To Distinctly Release miRNA and Growth Factors To Enrich Regulatory T Cells and Rescue Periodontal Bone Loss. ACS NANO 2018; 12:9785-9799. [PMID: 30141906 PMCID: PMC6205210 DOI: 10.1021/acsnano.7b08976] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In addition to T cells' roles in immune response and autoimmune diseases, certain types of T cells, called regulatory T cells (Tregs), play important roles in microenvironment modulation for resolution and tissue regeneration. However, there are currently few options available other than introducing more Tregs or immunosuppressive drugs to locally enrich Tregs. Herein, poly(l-lactic acid) (PLLA) nanofibrous spongy microspheres (NF-SMS), PLLA/polyethylene glycol (PEG) co-functionalized mesoporous silica nanoparticles (MSN), and poly(lactic acid- co-glycolic acid) microspheres (PLGA MS) are integrated into one multibiologic delivery vehicle for in situ Treg manipulation, where the MSNs and PLGA MS were utilized to distinctly release IL-2/TGF-β and miR-10a to locally recruit T cells and stimulate their differentiation into Tregs, while PLLA NF-SMS serve as an injectable scaffold for the adhesion and proliferation of these Tregs. In a mouse model of periodontitis, the injectable and biomolecule-delivering PLLA NF-SMS lead to Treg enrichment, expansion, and Treg-mediated immune therapy against bone loss. This system can potentially be utilized in a wide variety of other immune and regenerative therapies.
Collapse
Affiliation(s)
- Zhongning Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology; National Clinical Research Center for Oral Diseases; National Engineering Laboratory for Digital and Material Technology of Stomatology; Beijing Key Laboratory of Digital Stomatology, Beijing 100081, PR China
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Xin Chen
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhanpeng Zhang
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaojin Zhang
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Saunders
- Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology; National Clinical Research Center for Oral Diseases; National Engineering Laboratory for Digital and Material Technology of Stomatology; Beijing Key Laboratory of Digital Stomatology, Beijing 100081, PR China
- Corresponding Authors (Y. Zhou): , (P.X. Ma):
| | - Peter X. Ma
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
- Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, Department of Materials Sciences and Engineering, The University of Michigan, Ann Arbor, MI 48109, USA
- Corresponding Authors (Y. Zhou): , (P.X. Ma):
| |
Collapse
|
84
|
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
|
85
|
Hickey JW, Kosmides AK, Schneck JP. Engineering Platforms for T Cell Modulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:277-362. [PMID: 30262034 DOI: 10.1016/bs.ircmb.2018.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T cells are crucial contributors to mounting an effective immune response and increasingly the focus of therapeutic interventions in cancer, infectious disease, and autoimmunity. Translation of current T cell immunotherapies has been hindered by off-target toxicities, limited efficacy, biological variability, and high costs. As T cell therapeutics continue to develop, the application of engineering concepts to control their delivery and presentation will be critical for their success. Here, we outline the engineer's toolbox and contextualize it with the biology of T cells. We focus on the design principles of T cell modulation platforms regarding size, shape, material, and ligand choice. Furthermore, we review how application of these design principles has already impacted T cell immunotherapies and our understanding of T cell biology. Recent, salient examples from protein engineering, synthetic particles, cellular and genetic engineering, and scaffolds and surfaces are provided to reinforce the importance of design considerations. Our aim is to provide a guide for immunologists, engineers, clinicians, and the pharmaceutical sector for the design of T cell-targeting platforms.
Collapse
Affiliation(s)
- John W Hickey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa K Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan P Schneck
- Institute for NanoBiotechnology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
86
|
Gosselin EA, Eppler HB, Bromberg JS, Jewell CM. Designing natural and synthetic immune tissues. NATURE MATERIALS 2018; 17:484-498. [PMID: 29784994 PMCID: PMC6283404 DOI: 10.1038/s41563-018-0077-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 04/11/2018] [Indexed: 05/10/2023]
Abstract
Vaccines and immunotherapies have provided enormous improvements for public health, but there are fundamental disconnects between where most studies are performed-in cell culture and animal models-and the ultimate application in humans. Engineering immune tissues and organs, such as bone marrow, thymus, lymph nodes and spleen, could be instrumental in overcoming these hurdles. Fundamentally, designed immune tissues could serve as in vitro tools to more accurately study human immune function and disease, while immune tissues engineered for implantation as next-generation vaccines or immunotherapies could enable direct, on-demand control over generation and regulation of immune function. In this Review, we discuss recent interdisciplinary strategies that are merging materials science and immunology to create engineered immune tissues in vitro and in vivo. We also highlight the hurdles facing these approaches and the need for comparison to existing clinical options, relevant animal models, and other emerging technologies.
Collapse
Affiliation(s)
- Emily A Gosselin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Haleigh B Eppler
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Molecular and Cellular Biology, Biological Sciences Training Program, University of Maryland, College Park, MD, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Molecular and Cellular Biology, Biological Sciences Training Program, University of Maryland, College Park, MD, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, USA.
- United States Department of Veterans Affairs, Maryland VA Health Care System, Baltimore, MD, USA.
| |
Collapse
|
87
|
Tyagi P, Santos JL. Macromolecule nanotherapeutics: approaches and challenges. Drug Discov Today 2018; 23:1053-1061. [DOI: 10.1016/j.drudis.2018.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/09/2017] [Accepted: 01/04/2018] [Indexed: 01/29/2023]
|
88
|
Leng F, Liu F, Yang Y, Wu Y, Tian W. Strategies on Nanodiagnostics and Nanotherapies of the Three Common Cancers. NANOMATERIALS 2018; 8:nano8040202. [PMID: 29597315 PMCID: PMC5923532 DOI: 10.3390/nano8040202] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/18/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
The emergence of nanomedicine has enriched the knowledge and strategies of treating diseases, and especially some incurable diseases, such as cancers, acquired immune deficiency syndrome (AIDS), and neurodegenerative diseases. The application of nanoparticles in medicine is in the core of nanomedicine. Nanoparticles can be used in drug delivery for improving the uptake of poorly soluble drugs, targeted delivery to a specific site, and drug bioavailability. Early diagnosis of and targeted therapies for cancers can significantly improve patients' quality of life and extend patients' lives. The advantages of nanoparticles have given them a progressively important role in the nanodiagnosis and nanotherapy of common cancers. To provide a reference for the further application of nanoparticles, this review focuses on the recent development and application of nanoparticles in the early diagnosis and treatment of the three common cancers (lung cancer, liver cancer, and breast cancer) by using quantum dots, magnetic nanoparticles, and gold nanoparticles.
Collapse
Affiliation(s)
- Fan Leng
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Fang Liu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yongtao Yang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yu Wu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Weiqun Tian
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
89
|
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
|
90
|
|
91
|
Cheung AS, Zhang DK, Koshy ST, Mooney DJ. Scaffolds that mimic antigen-presenting cells enable ex vivo expansion of primary T cells. Nat Biotechnol 2018; 36:160-169. [PMID: 29334370 PMCID: PMC5801009 DOI: 10.1038/nbt.4047] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/01/2017] [Indexed: 12/22/2022]
Abstract
Therapeutic ex vivo T-cell expansion is limited by low rates and T-cell products of limited functionality. Here we describe a system that mimics natural antigen-presenting cells (APCs) and consists of a fluid lipid bilayer supported by mesoporous silica micro-rods. The lipid bilayer presents membrane-bound cues for T-cell receptor stimulation and costimulation, while the micro-rods enable sustained release of soluble paracrine cues. Using anti-CD3, anti-CD28, and interleukin-2, we show that the APC-mimetic scaffolds (APC-ms) promote two- to tenfold greater polyclonal expansion of primary mouse and human T cells compared with commercial expansion beads (Dynabeads). The efficiency of expansion depends on the density of stimulatory cues and the amount of material in the starting culture. Following a single stimulation, APC-ms enables antigen-specific expansion of rare cytotoxic T-cell subpopulations at a greater magnitude than autologous monocyte-derived dendritic cells after 2 weeks. APC-ms support over fivefold greater expansion of restimulated CD19 CAR-T cells than Dynabeads, with similar efficacy in a xenograft lymphoma model.
Collapse
Affiliation(s)
- Alexander S. Cheung
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| | - David K.Y. Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| | - Sandeep T. Koshy
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- The Wyss Institute for Biologically Inspired Engineering Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
92
|
Xie YQ, Wei L, Tang L. Immunoengineering with biomaterials for enhanced cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 10:e1506. [PMID: 29333729 DOI: 10.1002/wnan.1506] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/07/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has recently shown dramatic clinical success inducing durable response in patients of a wide variety of malignancies. Further improvement of the clinical outcome with immune related cancer treatment requests more exquisite manipulation of a patient's immune system with increased immunity against diseases while mitigating the toxicities. To meet this challenge, biomaterials applied to immunoengineering are being developed to achieve tissue- and/or cell-specific immunomodulation and thus could potentially enhance both the efficacy and safety of current cancer immunotherapies. Here, we review the recent advancement in the field of immunoengineering using biomaterials and their applications in promoting different modalities of cancer immunotherapies, with focus on cell-, antibody-, immunomodulator-, and gene-based immune related treatments and their combinations with conventional therapies. Challenges and opportunities are discussed in applying biomaterials engineering strategies in the development of future cancer immunotherapies. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lixia Wei
- Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute of Materials Science & Engineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
93
|
Zhang T, Li M, Wang X, Zhou Z, Yuan W, Ma J. Facile synthesis of polylactide coarse microspheres as artificial antigen-presenting cells. Chem Commun (Camb) 2018; 54:11356-11359. [DOI: 10.1039/c8cc04958k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Magnetic poly(l-lactide) coarse microspheres as artificial antigen-presenting cells were synthesized via simple chemical etching and antibody immobilization.
Collapse
Affiliation(s)
- Tong Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Min Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials
- Tianjin
- China
| | - Xiaotong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials
- Tianjin
- China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Jie Ma
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| |
Collapse
|
94
|
Sidhom JW, Bessell CA, Havel JJ, Kosmides A, Chan TA, Schneck JP. ImmunoMap: A Bioinformatics Tool for T-cell Repertoire Analysis. Cancer Immunol Res 2017; 6:151-162. [PMID: 29263161 DOI: 10.1158/2326-6066.cir-17-0114] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 09/12/2017] [Accepted: 12/18/2017] [Indexed: 11/16/2022]
Abstract
Despite a dramatic increase in T-cell receptor (TCR) sequencing, few approaches biologically parse the data in a fashion that both helps yield new information about immune responses and may guide immunotherapeutic interventions. To address this issue, we developed a method, ImmunoMap, that utilizes a sequence analysis approach inspired by phylogenetics to examine TCR repertoire relatedness. ImmunoMap analysis of the CD8 T-cell response to self-antigen (Kb-TRP2) or to a model foreign antigen (Kb-SIY) in naïve and tumor-bearing B6 mice showed differences in the T-cell repertoire of self- versus foreign antigen-specific responses, potentially reflecting immune pressure by the tumor, and also detected lymphoid organ-specific differences in TCR repertoires. When ImmunoMap was used to analyze clinical trial data of tumor-infiltrating lymphocytes from patients being treated with anti-PD-1, ImmunoMap, but not standard TCR sequence analyses, revealed a clinically predicative signature in pre- and posttherapy samples. Cancer Immunol Res; 6(2); 151-62. ©2017 AACR.
Collapse
Affiliation(s)
- John-William Sidhom
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Catherine A Bessell
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan J Havel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alyssa Kosmides
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonathan P Schneck
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Nanobiotechnology, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
95
|
Gammon JM, Dold NM, Jewell CM. Improving the clinical impact of biomaterials in cancer immunotherapy. Oncotarget 2017; 7:15421-43. [PMID: 26871948 PMCID: PMC4941251 DOI: 10.18632/oncotarget.7304] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/29/2016] [Indexed: 12/20/2022] Open
Abstract
Immunotherapies for cancer have progressed enormously over the past few decades, and hold great promise for the future. The successes of these therapies, with some patients showing durable and complete remission, demonstrate the power of harnessing the immune system to eradicate tumors. However, the effectiveness of current immunotherapies is limited by hurdles ranging from immunosuppressive strategies employed by tumors, to inadequate specificity of existing therapies, to heterogeneity of disease. Further, the vast majority of approved immunotherapies employ systemic delivery of immunomodulators or cells that make addressing some of these challenges more difficult. Natural and synthetic biomaterials–such as biocompatible polymers, self-assembled lipid particles, and implantable biodegradable devices–offer unique potential to address these hurdles by harnessing the benefits of therapeutic targeting, tissue engineering, co-delivery, controlled release, and sensing. However, despite the enormous investment in new materials and nanotechnology, translation of these ideas to the clinic is still an uncommon outcome. Here we review the major challenges facing immunotherapies and discuss how the newest biomaterials and nanotechnologies could help overcome these challenges to create new clinical options for patients.
Collapse
Affiliation(s)
- Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Neil M Dold
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.,Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| |
Collapse
|
96
|
Shen H, Sun T, Hoang HH, Burchfield JS, Hamilton GF, Mittendorf EA, Ferrari M. Enhancing cancer immunotherapy through nanotechnology-mediated tumor infiltration and activation of immune cells. Semin Immunol 2017; 34:114-122. [PMID: 28947107 PMCID: PMC5705528 DOI: 10.1016/j.smim.2017.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy has become arguably the most promising advancement in cancer research and therapy in recent years. The efficacy of cancer immunotherapy is critically dependent on specific physiological and physical processes - collectively referred to as transport barriers - including the activation of T cells by antigen presenting cells, T cells migration to and penetration into the tumor microenvironment, and movement of nutrients and other immune cells through the tumor microenvironment. Nanotechnology-based approaches have great potential to help overcome these transport barriers. In this review, we discuss the ways that nanotechnology is being leveraged to improve the efficacy and potency of various cancer immunotherapies.
Collapse
Affiliation(s)
- Haifa Shen
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tong Sun
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Hanh H Hoang
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Jana S Burchfield
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Gillian F Hamilton
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mauro Ferrari
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
97
|
Shen C, Xu T, Wu Y, Li X, Xia L, Wang W, Shahzad KA, Zhang L, Wan X, Qiu J. Frequency and reactivity of antigen-specific T cells were concurrently measured through the combination of artificial antigen-presenting cell, MACS and ELISPOT. Sci Rep 2017; 7:16400. [PMID: 29180767 PMCID: PMC5703716 DOI: 10.1038/s41598-017-16549-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/14/2017] [Indexed: 01/24/2023] Open
Abstract
Conventional peptide-major histocompatibility complex (pMHC) multimer staining, intracellular cytokine staining, and enzyme-linked immunospot (ELISPOT) assay cannot concurrently determine the frequency and reactivity of antigen-specific T cells (AST) in a single assay. In this report, pMHC multimer, magnetic-activated cell sorting (MACS), and ELISPOT techniques have been integrated into a micro well by coupling pMHC multimers onto cell-sized magnetic beads to characterize AST cell populations in a 96-well microplate which pre-coated with cytokine-capture antibodies. This method, termed AAPC-microplate, allows the enumeration and local cytokine production of AST cells in a single assay without using flow cytometry or fluorescence intensity scanning, thus will be widely applicable. Here, ovalbumin257-264-specific CD8+ T cells from OT-1 T cell receptor (TCR) transgenic mice were measured. The methodological accuracy, specificity, reproducibility, and sensitivity in enumerating AST cells compared well with conventional pMHC multimer staining. Furthermore, the AAPC-microplate was applied to detect the frequency and reactivity of Hepatitis B virus (HBV) core antigen18-27- and surface antigen183-191-specific CD8+ T cells for the patients, and was compared with conventional method. This method without the need of high-end instruments may facilitate the routine analysis of patient-specific cellular immune response pattern to a given antigen in translational studies.
Collapse
Affiliation(s)
- Chuanlai Shen
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China.
| | - Tao Xu
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - You Wu
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Xiaoe Li
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Lingzhi Xia
- Department of Laboratory Medicine, Nanjing KingMed Diagnostics Company Limited, Nanjing, Jiangsu, China
| | - Wei Wang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Lei Zhang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Xin Wan
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Jie Qiu
- Division of Infectious Diseases, Second Hospital of Nanjing, Affiliated Second Hospital of Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
98
|
Hickey JW, Vicente FP, Howard GP, Mao HQ, Schneck JP. Biologically Inspired Design of Nanoparticle Artificial Antigen-Presenting Cells for Immunomodulation. NANO LETTERS 2017; 17:7045-7054. [PMID: 28994285 PMCID: PMC6709596 DOI: 10.1021/acs.nanolett.7b03734] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Particles engineered to engage and interact with cell surface ligands and to modulate cells can be harnessed to explore basic biological questions as well as to devise cellular therapies. Biology has inspired the design of these particles, such as artificial antigen-presenting cells (aAPCs) for use in immunotherapy. While much has been learned about mimicking antigen presenting cell biology, as we decrease the size of aAPCs to the nanometer scale, we need to extend biomimetic design to include considerations of T cell biology-including T-cell receptor (TCR) organization. Here we describe the first quantitative analysis of particle size effect on aAPCs with both Signals 1 and 2 based on T cell biology. We show that aAPCs, larger than 300 nm, activate T cells more efficiently than smaller aAPCs, 50 nm. The 50 nm aAPCs require saturating doses or require artificial magnetic clustering to activate T cells. Increasing ligand density alone on the 50 nm aAPCs did not increase their ability to stimulate CD8+ T cells, confirming the size-dependent phenomenon. These data support the need for multireceptor ligation and activation of T-cell receptor (TCR) nanoclusters of similar sizes to 300 nm aAPCs. Quantitative analysis and modeling of a nanoparticle system provides insight into engineering constraints of aAPCs for T cell immunotherapy applications and offers a case study for other cell-modulating particles.
Collapse
Affiliation(s)
- John W. Hickey
- Department of Biomedical Engineering, School of Medicine
- Institute for Cell Engineering, School of Medicine
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
| | | | - Gregory P. Howard
- Department of Biomedical Engineering, School of Medicine
- Institute for Nanobiotechnology
| | - Hai-Quan Mao
- Translational Tissue Engineering Center
- Institute for Nanobiotechnology
- Department of Materials Science and Engineering, Whiting School of Engineering
| | - Jonathan P. Schneck
- Institute for Cell Engineering, School of Medicine
- Department of Pathology, School of Medicine
- Institute for Nanobiotechnology
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Corresponding Author: . Phone: 410-614-4589
| |
Collapse
|
99
|
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
|
100
|
Wang C, Sun W, Ye Y, Bomba HN, Gu Z. Bioengineering of Artificial Antigen Presenting Cells and Lymphoid Organs. Theranostics 2017; 7:3504-3516. [PMID: 28912891 PMCID: PMC5596439 DOI: 10.7150/thno.19017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/24/2017] [Indexed: 12/12/2022] Open
Abstract
The immune system protects the body against a wide range of infectious diseases and cancer by leveraging the efficiency of immune cells and lymphoid organs. Over the past decade, immune cell/organ therapies based on the manipulation, infusion, and implantation of autologous or allogeneic immune cells/organs into patients have been widely tested and have made great progress in clinical applications. Despite these advances, therapy with natural immune cells or lymphoid organs is relatively expensive and time-consuming. Alternatively, biomimetic materials and strategies have been applied to develop artificial immune cells and lymphoid organs, which have attracted considerable attentions. In this review, we survey the latest studies on engineering biomimetic materials for immunotherapy, focusing on the perspectives of bioengineering artificial antigen presenting cells and lymphoid organs. The opportunities and challenges of this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hunter N. Bomba
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|