1
|
Lin JN, Kuan CY, Chang CT, Chen ZY, Kuo WT, Lin J, Lin YY, Yang IH, Lin FH. High-throughput proliferation and activation of NK-92MI cell spheroids via a homemade one-step closed bioreactor in pseudostatic cultures for immunocellular therapy. J Biol Eng 2024; 18:65. [PMID: 39533411 PMCID: PMC11555828 DOI: 10.1186/s13036-024-00461-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The NK-92MI cell line has displayed significant promise in clinical trials for cancer treatment. However, challenges persist in obtaining sufficient cell quantities and achieving optimal cytotoxicity. The proliferation of natural killer (NK) cells involves the formation of cell aggregates, but excessively large aggregates can impede nutrient and waste transport, leading to reduced cell survival rates. In this study, a custom bioreactor was designed to mimic pseudostatic culture conditions by integrating brief mechanical rotation during a 6-h static culture period. This method aimed to achieve an optimal aggregate size while improving cell viability. The findings revealed a 144-fold expansion of 3D NK-92MI cell aggregates, reaching an ideal size of 80-150 µm, significantly increasing both cell proliferation and survival rates. After 14 days of culture, the NK-92MI cells maintained their phenotype during the subsequent phase of cell activation. Moreover, these cells presented elevated levels of IFN-γ expression after IL-18 activation, resulting in enhanced NK cell-mediated cytotoxicity against K562 cells. This innovative strategy, which uses a closed suspension-based culture system, presents a promising approach for improving cell expansion and activation techniques in immunocellular therapy.
Collapse
Affiliation(s)
- Jhih-Ni Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan
| | - Che-Yung Kuan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan
| | - Chia-Ting Chang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan
- National Chung Hsing University, Taichung, Taiwan
| | - Zhi-Yu Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan
| | - Wei-Ting Kuo
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan
| | - Jason Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan
| | - Yu-Ying Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan
- National Chung Hsing University, Taichung, Taiwan
| | - I-Hsuan Yang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan.
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan.
| | - Feng-Huei Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli, 35053, Taiwan.
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 49, Fanglan Rd, Taipei, 10672, Taiwan.
| |
Collapse
|
2
|
Chen ZP, Zeng WJ, Lei YM, Liang WB, Yang X, Yuan R, Yang C, Zhuo Y. Engineering of a Multi-Modular DNA Nanodevice for Spatioselective Imaging and Evaluation of NK Cell-Mediated Cancer Immunotherapy. Angew Chem Int Ed Engl 2024:e202414064. [PMID: 39375853 DOI: 10.1002/anie.202414064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
Granzyme A (GzmA) secreted by natural killer (NK) cells has garnered considerable interest as a biomarker to evaluate the efficacy of cancer immunotherapy. However, current methodologies to selectively monitor the spatial distribution of GzmA in cancer cells during NK cell-targeted therapy are extremely challenging, primarily due to the existence of diverse cell populations, the low levels of GzmA expression, and the limited availability of GzmA probes. Herein we develop a multi-modular, structurally-ordered DNA nanodevice for evaluating NK cell-mediated cancer immunotherapy (MODERN), that permits spatioselective imaging of GzmA in cancer cells through GzmA-induced apurinic/apyrimidinic endonuclease 1 (APE1) inactivation. The MODERN incorporates multiple functional modules, including an APE1-gated recognition module, a photo-activated amplification module, an aptamer-mediated tumor-target module, and a polycatenane DNA module, enabling improved sensitivity and specificity towards intracellular GzmA. The MODERN was activated (on) in cancer cells due to the overexpression of APE1, whereas it remained silent (off) in the NK-treated cancer cells owing to the GzmA-induced APE1 inactivation. Furthermore, we demonstrated that GzmA-induced APE1 inactivation blocks the cellular repair of target cells, resulting in efficient cell death. This MODERN that relies on the specific inactivation of APE1 by GzmA should be beneficial for evaluating the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Zhao-Peng Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Wei-Jia Zeng
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Yan-Mei Lei
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Chongqing, 400715, China
| | - Wen-Bin Liang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Xia Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, China
| |
Collapse
|
3
|
Koo D, Mao Z, Dimatteo R, Noguchi M, Tsubamoto N, McLaughlin J, Tran W, Lee S, Cheng D, de Rutte J, Burton Sojo G, Witte ON, Di Carlo D. Defining T cell receptor repertoires using nanovial-based binding and functional screening. Proc Natl Acad Sci U S A 2024; 121:e2320442121. [PMID: 38536748 PMCID: PMC10998554 DOI: 10.1073/pnas.2320442121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/27/2024] [Indexed: 04/08/2024] Open
Abstract
The ability to selectively bind to antigenic peptides and secrete effector molecules can define rare and low-affinity populations of cells with therapeutic potential in emerging T cell receptor (TCR) immunotherapies. We leverage cavity-containing hydrogel microparticles, called nanovials, each coated with peptide-major histocompatibility complex (pMHC) monomers to isolate antigen-reactive T cells. T cells are captured and activated by pMHCs inducing the secretion of effector molecules including IFN-γ and granzyme B that are accumulated on nanovials, allowing sorting based on both binding and function. The TCRs of sorted cells on nanovials are sequenced, recovering paired αβ-chains using microfluidic emulsion-based single-cell sequencing. By labeling nanovials having different pMHCs with unique oligonucleotide-barcodes and secretions with oligo-barcoded detection antibodies, we could accurately link TCR sequences to specific targets and rank each TCR based on the corresponding cell's secretion level. Using the technique, we identified an expanded repertoire of functional TCRs targeting viral antigens with high specificity and found rare TCRs with activity against cancer-specific splicing-enhanced epitopes.
Collapse
Affiliation(s)
- Doyeon Koo
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA90095
| | - Miyako Noguchi
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Natalie Tsubamoto
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Sohyung Lee
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA90095
| | - Donghui Cheng
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
| | - Joseph de Rutte
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Partillion Bioscience, Pasadena, CA91107
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA90095
- Molecular Biology Institute, University of California, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Partillion Bioscience, Pasadena, CA91107
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA90095
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA90095
- California NanoSystems Institute, Los Angeles, CA90095
| |
Collapse
|
4
|
Terada Y, Obara A, Briones JC, Luo X, Espulgar WV, Saito M, Takamatsu H, Tamiya E. Development of Nano-Micro Fused LSPR Chip for In Situ Single-Cell Secretion Analysis. MICROMACHINES 2023; 14:1404. [PMID: 37512715 PMCID: PMC10384685 DOI: 10.3390/mi14071404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023]
Abstract
Single-cell analysis has become increasingly important in uncovering cell heterogeneity, which has great implications in medicine and biology for a deep understanding of cell characteristics. Owing to its significance, it is vital to create novel devices that can reveal special or unique cells. In this work, we developed a single-cell secretion detection chip consisting of microwells that can trap single cells. Each well is surrounded by Au nanopillars capable of localized surface plasmon resonance (LSPR) measurement. Using microfabrication and nanofabrication techniques, Au nanopillar and microwell structures were fabricated on a COP film. The Au nanopillar was modified with IL-6 antibodies for the direct detection of single-cell secreted IL-6 via LSPR absorbance peak shift. Specific IL-6 detection was successfully demonstrated using a null and IL-6 oversecreting Jurkat cell. A high single-cell trapping efficiency of over 80% was also achieved. Overall, the development of this single-cell secretion detection chip with a simple LSPR measurement setup represents a significant development in the field of cell biology and immunology, providing researchers with a powerful tool for studying individual cells and their secreted cytokines, and is useful for point-of-care testing (POCT) diagnostics.
Collapse
Affiliation(s)
- Yuhei Terada
- Environmental Management Research Institute (EMRI), Department of Energy and Environment, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8569, Ibaraki, Japan
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita 565-0871, Osaka, Japan
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita 565-0871, Osaka, Japan
| | - Ain Obara
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita 565-0871, Osaka, Japan
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita 565-0871, Osaka, Japan
| | - Jonathan Campos Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Osaka, Japan
| | - Xi Luo
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita 565-0871, Osaka, Japan
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita 565-0871, Osaka, Japan
| | - Wilfred Villariza Espulgar
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita 565-0871, Osaka, Japan
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Suita 565-0871, Osaka, Japan
| | - Masato Saito
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita 565-0871, Osaka, Japan
- Laboratory of Autoimmune Diseases, Department of Clinical Research Center for Autoimmune Diseases, NHO Osaka Minami Medical Center, Kawachinagano 586-8521, Osaka, Japan
| | - Eiichi Tamiya
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Suita 565-0871, Osaka, Japan
- SANKEN, Osaka University, Ibaraki 567-0047, Osaka, Japan
| |
Collapse
|
5
|
Briones JC, Espulgar WV, Koyama S, Takamatsu H, Saito M, Tamiya E. A High-Throughput Single-Cell Assay on a Valve-Based Microfluidic Platform Applied to Protein Quantification, Immune Response Monitoring, and Drug Discovery. Methods Mol Biol 2023; 2689:119-142. [PMID: 37430051 DOI: 10.1007/978-1-0716-3323-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The use of microfluidic technology in single-cell assay has shown potential in biomedical applications like protein quantification, immune response monitoring, and drug discovery. Because of the details of information that can be obtained at single-cell resolution, the single-cell assay has been applied to tackle challenging issues such as cancer treatment. Information like the levels of protein expression, cellular heterogeneity, and unique behaviors within subsets are very important in the biomedical field. For a single-cell assay system, a high-throughput platform that can do on-demand media exchange and real-time monitoring is advantageous in single-cell screening and profiling. In this work, a high-throughput valve-based device is presented, its use in single-cell assay, particularly in protein quantification and surface-marker analysis, and its potential application to immune response monitoring and drug discovery are laid down in detail.
Collapse
Affiliation(s)
- Jonathan C Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Wilfred V Espulgar
- Department of Physics, College of Science, De La Salle University, Manila, Philippines
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masato Saito
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- National Institute of Advanced Industrial Science and Technology, PhotoBIO Open Innovation Laboratory, Osaka, Japan
| | - Eiichi Tamiya
- National Institute of Advanced Industrial Science and Technology, PhotoBIO Open Innovation Laboratory, Osaka, Japan.
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan.
| |
Collapse
|
6
|
Xu L, Liu N, Zhan W, Deng Y, Chen Z, Liu X, Gao G, Chen Q, Liu Z, Liang G. Granzyme B Turns Nanoparticle Fluorescence "On" for Imaging Cytotoxic T Lymphocyte Activity in Vivo. ACS NANO 2022; 16:19328-19334. [PMID: 36282211 DOI: 10.1021/acsnano.2c08896] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are important immune cells, and their activation is a key step for cancer immunotherapy. Precise evaluation of CTL activity in vivo provides a powerful tool for monitoring cancer-immunotherapeutic outcomes, yet it faces tremendous challenges. Herein, by rationally designing a near-infrared (NIR) fluorescence probe Cys(StBu)-Ile-Glu-Phe-Asp-Lys(Cy5.5)-CBT (Cy5.5-CBT) and employing a reduction-instructed CBT-Cys click condensation reaction, we developed the fluorescence "dual quenched" nanoparticles Cy5.5-CBT-NPs for imaging of granzyme B (GraB), a biomarker tightly associated with the tumoricidal activity of CTLs. Upon GraB cleavage, Cy5.5-CBT-NPs disassembled, subtly turning the fluorescence signal "on". With this fluorescence "turn-on" property, Cy5.5-CBT-NPs enabled sensitive and real-time monitoring of GraB-mediated CTL responses against cancer cells in vitro. Animal experiments demonstrated that, at 16 h post injection, the fluorescence imaging signal of Cy5.5-CBT-NPs showed a 3.1-fold increase on the tumor sites of mice treated by an immune-activating drug S-(2-boronoethyl)-L-cysteine hydrochloride. We envision that Cy5.5-CBT-NPs may provide a powerful tool for noninvasive and sensitive evaluation of immunotherapeutic efficacy of cancer in the near future.
Collapse
Affiliation(s)
- Lingling Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Nanhui Liu
- Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Wenjun Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Yu Deng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Zhaoxia Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Ge Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| | - Qian Chen
- Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Zhuang Liu
- Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou, Nanjing 210096, China
| |
Collapse
|
7
|
Ma X, Zhang MJ, Wang J, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Emerging Biomaterials Imaging Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204034. [PMID: 35728795 DOI: 10.1002/adma.202204034] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy is one of the most promising clinical modalities for the treatment of malignant tumors and has shown excellent therapeutic outcomes in clinical settings. However, it continues to face several challenges, including long treatment cycles, high costs, immune-related adverse events, and low response rates. Thus, it is critical to predict the response rate to immunotherapy by using imaging technology in the preoperative and intraoperative. Here, the latest advances in nanosystem-based biomaterials used for predicting responses to immunotherapy via the imaging of immune cells and signaling molecules in the immune microenvironment are comprehensively summarized. Several imaging methods, such as fluorescence imaging, magnetic resonance imaging, positron emission tomography imaging, ultrasound imaging, and photoacoustic imaging, used in immune predictive imaging, are discussed to show the potential of nanosystems for distinguishing immunotherapy responders from nonresponders. Nanosystem-based biomaterials aided by various imaging technologies are expected to enable the effective prediction and diagnosis in cases of tumors, inflammation, and other public diseases.
Collapse
Affiliation(s)
- Xianbin Ma
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jingting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Tian Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
8
|
Li Y, Fan H, Ding J, Xu J, Liu C, Wang H. Microfluidic devices: The application in TME modeling and the potential in immunotherapy optimization. Front Genet 2022; 13:969723. [PMID: 36159996 PMCID: PMC9493116 DOI: 10.3389/fgene.2022.969723] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
With continued advances in cancer research, the crucial role of the tumor microenvironment (TME) in regulating tumor progression and influencing immunotherapy outcomes has been realized over the years. A series of studies devoted to enhancing the response to immunotherapies through exploring efficient predictive biomarkers and new combination approaches. The microfluidic technology not only promoted the development of multi-omics analyses but also enabled the recapitulation of TME in vitro microfluidic system, which made these devices attractive across studies for optimization of immunotherapy. Here, we reviewed the application of microfluidic systems in modeling TME and the potential of these devices in predicting and monitoring immunotherapy effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Huiyu Wang
- *Correspondence: Chaoying Liu, ; Huiyu Wang,
| |
Collapse
|
9
|
Zhang J, Tavakoli H, Ma L, Li X, Han L, Li X. Immunotherapy discovery on tumor organoid-on-a-chip platforms that recapitulate the tumor microenvironment. Adv Drug Deliv Rev 2022; 187:114365. [PMID: 35667465 DOI: 10.1016/j.addr.2022.114365] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/17/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has achieved remarkable success over the past decade by modulating patients' own immune systems and unleashing pre-existing immunity. However, only a minority of cancer patients across different cancer types are able to benefit from immunotherapy treatment; moreover, among those small portions of patients with response, intrinsic and acquired resistance remains a persistent challenge. Because the tumor microenvironment (TME) is well recognized to play a critical role in tumor initiation, progression, metastasis, and the suppression of the immune system and responses to immunotherapy, understanding the interactions between the TME and the immune system is a pivotal step in developing novel and efficient cancer immunotherapies. With unique features such as low reagent consumption, dynamic and precise fluid control, versatile structures and function designs, and 3D cell co-culture, microfluidic tumor organoid-on-a-chip platforms that recapitulate key factors of the TME and the immune contexture have emerged as innovative reliable tools to investigate how tumors regulate their TME to counteract antitumor immunity and the mechanism of tumor resistance to immunotherapy. In this comprehensive review, we focus on recent advances in tumor organoid-on-a-chip platforms for studying the interaction between the TME and the immune system. We first review different factors of the TME that recent microfluidic in vitro systems reproduce to generate advanced tools to imitate the crosstalk between the TME and the immune system. Then, we discuss their applications in the assessment of different immunotherapies' efficacy using tumor organoid-on-a-chip platforms. Finally, we present an overview and the outlook of engineered microfluidic platforms in investigating the interactions between cancer and immune systems, and the adoption of patient-on-a-chip models in clinical applications toward personalized immunotherapy.
Collapse
Affiliation(s)
- Jie Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Hamed Tavakoli
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Lei Ma
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA
| | - Xiaochun Li
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Lichun Han
- Xi'an Daxing Hospital, Xi'an 710016, China
| | - XiuJun Li
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA; Border Biomedical Research Center, Forensic Science, & Environmental Science and Engineering, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, USA.
| |
Collapse
|
10
|
Ramirez A, Amosu M, Lee P, Maisel K. Microfluidic systems to study tissue barriers to immunotherapy. Drug Deliv Transl Res 2021; 11:2414-2429. [PMID: 34215998 PMCID: PMC9059778 DOI: 10.1007/s13346-021-01016-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 10/20/2022]
Abstract
Immunotherapies have been heavily explored in the last decade, ranging from new treatments for cancer to allergic diseases. These therapies target the immune system, a complex organ system consisting of tissues with intricate structures and cells with a multitude of functions. To better understand immune functions and develop better therapeutics, many cellular and 2-dimensional (2D) tissue models have been developed. However, research has demonstrated that the 3-dimensional (3D) tissue structure can significantly affect cellular functions, and this is not recapitulated by more traditional 2D models. Microfluidics has been used to design 3D tissue models that allow for intricate arrangements of cells and extracellular spaces, thus allowing for more physiologically relevant in vitro model systems. Here, we summarize the multitude of microfluidic devices designed to study the immune system with the ultimate goal to improve existing and design new immunotherapies. We have included models of the different immune organs, including bone marrow and lymph node (LN), models of immunity in diseases such as cancer and inflammatory bowel disease, and therapeutic models to test or engineer new immune-modulatory treatments. We particularly emphasize research on how microfluidic devices are used to better understand different physiological states and how interactions within the immune microenvironment can influence the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Ann Ramirez
- Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Mayowa Amosu
- Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Priscilla Lee
- Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Katharina Maisel
- Department of Bioengineering, University of Maryland, College Park, MD, USA.
| |
Collapse
|
11
|
Shi H, Gong Y, Liang Q, Li J, Xiang Y, Li G. Target-Initiated Great Change in Electrochemical Steric Hindrance for an Assay of Granzyme B Activity. Anal Chem 2021; 93:13382-13388. [PMID: 34549940 DOI: 10.1021/acs.analchem.1c03188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To improve long-term graft patient outcomes and develop more effective antirejection therapies, noninvasive monitoring of acute cellular rejection (ACR) after organ transplantation is urgently needed. As a biomarker of ACR, Granzyme B (GrB) is expected to be applied in the noninvasive monitoring of ACR. Herein, we have developed a method for detecting the GrB activity based on the target-initiated great change in electrochemical steric hindrance by designing a nanoprobe. The nanoprobe is prepared by conjugating a specific peptide, which is responsive to GrB cleavage activity, to gold nanoparticles (AuNPs). Meanwhile, a piece of DNA sequence with G-quadruplex (G4) is attached at the distal end of the peptide. Upon exposure to GrB, the peptide substrate is cleaved to eliminate the steric hindrance between inter-nanoprobes as well as nanoprobe and DNA tetrahedron (TDN), allowing the released DNA strand to hybridize with TDN, giving sensitive signal output. The method can also be used to detect GrB activity in complex biological settings, so it has a great potential for monitoring GrB activity in the blood or urine of graft patients.
Collapse
Affiliation(s)
- Hai Shi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Youjing Gong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Qizhi Liang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jinlong Li
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, P. R. China
| | - Yang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.,Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
12
|
Briones J, Espulgar W, Koyama S, Takamatsu H, Tamiya E, Saito M. The future of microfluidics in immune checkpoint blockade. Cancer Gene Ther 2021; 28:895-910. [PMID: 33110208 DOI: 10.1038/s41417-020-00248-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 01/30/2023]
Abstract
Recent advances in microfluidic techniques have enabled researchers to study sensitivities to immune checkpoint therapy, to determine patients' response to particular antibody treatment. Utilization of this technology is helpful in antibody discovery and in the design of personalized medicine. A variety of microfluidic approaches can provide several functions in processes such as immunologic, genomic, and/or transcriptomic analysis with the aim of improving the efficacy and coverage of immunotherapy, particularly immune checkpoint blockade (ICB). To achieve this requires researchers to overcome the challenges in the current state of the technology. This review looks into the advancements in microfluidic technologies applied to researches on immune checkpoint blockade treatment and its potential shift from proof-of-principle stage to clinical application.
Collapse
Affiliation(s)
- Jonathan Briones
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Wilfred Espulgar
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shohei Koyama
- Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hyota Takamatsu
- Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Eiichi Tamiya
- AIST PhotoBIO-OIL, Osaka University, Suita, Osaka, 565-0871, Japan.,The Institute of Scientific and Industrial Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masato Saito
- Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan. .,AIST PhotoBIO-OIL, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
13
|
Three-Dimensional Culture Models to Study Innate Anti-Tumor Immune Response: Advantages and Disadvantages. Cancers (Basel) 2021; 13:cancers13143417. [PMID: 34298630 PMCID: PMC8303518 DOI: 10.3390/cancers13143417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Several approaches have shown that the immune response against tumors strongly affects patients' clinical outcome. Thus, the study of anti-tumor immunity is critical to understand and potentiate the mechanisms underlying the elimination of tumor cells. Natural killer (NK) cells are members of innate immunity and represent powerful anti-tumor effectors, able to eliminate tumor cells without a previous sensitization. Thus, the study of their involvement in anti-tumor responses is critical for clinical translation. This analysis has been performed in vitro, co-incubating NK with tumor cells and quantifying the cytotoxic activity of NK cells. In vivo confirmation has been applied to overcome the limits of in vitro testing, however, the innate immunity of mice and humans is different, leading to discrepancies. Different activating receptors on NK cells and counter-ligands on tumor cells are involved in the antitumor response, and innate immunity is strictly dependent on the specific microenvironment where it takes place. Thus, three-dimensional (3D) culture systems, where NK and tumor cells can interact in a tissue-like architecture, have been created. For example, tumor cell spheroids and primary organoids derived from several tumor types, have been used so far to analyze innate immune response, replacing animal models. Herein, we briefly introduce NK cells and analyze and discuss in detail the properties of 3D tumor culture systems and their use for the study of tumor cell interactions with NK cells.
Collapse
|
14
|
A design and optimization of a high throughput valve based microfluidic device for single cell compartmentalization and analysis. Sci Rep 2021; 11:12995. [PMID: 34155296 PMCID: PMC8217553 DOI: 10.1038/s41598-021-92472-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/08/2021] [Indexed: 12/04/2022] Open
Abstract
The need for high throughput single cell screening platforms has been increasing with advancements in genomics and proteomics to identify heterogeneity, unique cell subsets or super mutants from thousands of cells within a population. For real-time monitoring of enzyme kinetics and protein expression profiling, valve-based microfluidics or pneumatic valving that can compartmentalize single cells is advantageous by providing on-demand fluid exchange capability for several steps in assay protocol and on-chip culturing. However, this technique is throughput limited by the number of compartments in the array. Thus, one big challenge lies in increasing the number of microvalves to several thousand that can be actuated in the microfluidic device to confine enzymes and substrates in picoliter volumes. This work explores the design and optimizations done on a microfluidic platform to achieve high-throughput single cell compartmentalization as applied to single-cell enzymatic assay for protein expression quantification. Design modeling through COMSOL Multiphysics was utilized to determine the circular microvalve’s optimized parameters, which can close thousands of microchambers in an array at lower sealing pressure. Multiphysical modeling results demonstrated the relationships of geometry, valve dimensions, and sealing pressure, which were applied in the fabrication of a microfluidic device comprising of up to 5000 hydrodynamic traps and corresponding microvalves. Comparing the effects of geometry, actuation media and fabrication technique, a sealing pressure as low as 0.04 MPa was achieved. Applying to single cell enzymatic assay, variations in granzyme B activity in Jurkat and human PBMC cells were observed. Improvement in the microfluidic chip’s throughput is significant in single cell analysis applications, especially in drug discovery and treatment personalization.
Collapse
|
15
|
Maulana TI, Kromidas E, Wallstabe L, Cipriano M, Alb M, Zaupa C, Hudecek M, Fogal B, Loskill P. Immunocompetent cancer-on-chip models to assess immuno-oncology therapy. Adv Drug Deliv Rev 2021; 173:281-305. [PMID: 33798643 DOI: 10.1016/j.addr.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The advances in cancer immunotherapy come with several obstacles, limiting its widespread use and benefits so far only to a small subset of patients. One of the underlying challenges remains to be the lack of representative nonclinical models that translate to human immunity and are able to predict clinical efficacy and safety outcomes. In recent years, immunocompetent Cancer-on-Chip models emerge as an alternative human-based platform that enables the integration and manipulation of complex tumor microenvironment. In this review, we discuss novel opportunities offered by Cancer-on-Chip models to advance (mechanistic) immuno-oncology research, ranging from design flexibility to multimodal analysis approaches. We then exemplify their (potential) applications for the research and development of adoptive cell therapy, immune checkpoint therapy, cytokine therapy, oncolytic virus, and cancer vaccines.
Collapse
|
16
|
The Present and Future Role of Microfluidics for Protein and Peptide-Based Therapeutics and Diagnostics. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11094109] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The implementation of peptide-based molecules within the medical field has vast potential, owing to their unique nature and predictable physicochemical profiles. However, peptide therapeutic usage is hindered by delivery-related challenges, meaning that their formulations must be altered to overcome these limitations. This process could be propelled by applying microfluidics (MFs) due to its highly controllable and adaptable attributes; however, therapeutic research within this field is extremely limited. Peptides possess multifunctional roles within therapeutic formulations, ranging from enhancing target specificity to acting as the active component of the medicine. Diagnostically, MFs are well explored in the field of peptides, as MFs provide an unsullied platform to provide fast yet accurate examinations. The capacity to add attributes, such as integrated sensors and microwells, to the MF chip, only enhances the attractiveness of MFs as a diagnostic platform. The structural individuality of peptides makes them prime candidates for diagnostic purposes, for example, antigen detection and isolation. Therefore, this review provides a useful insight into the current applications of MFs for peptide-based therapy and diagnostics and highlights potential gaps in the field that are yet to be explored or optimized.
Collapse
|
17
|
Parlato S, Grisanti G, Sinibaldi G, Peruzzi G, Casciola CM, Gabriele L. Tumor-on-a-chip platforms to study cancer-immune system crosstalk in the era of immunotherapy. LAB ON A CHIP 2021; 21:234-253. [PMID: 33315027 DOI: 10.1039/d0lc00799d] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Immunotherapy is a powerful therapeutic approach able to re-educate the immune system to fight cancer. A key player in this process is the tumor microenvironment (TME), which is a dynamic entity characterized by a complex array of tumor and stromal cells as well as immune cell populations trafficking to the tumor site through the endothelial barrier. Recapitulating these multifaceted dynamics is critical for studying the intimate interactions between cancer and the immune system and to assess the efficacy of emerging immunotherapies, such as immune checkpoint inhibitors (ICIs) and adoptive cell-based products. Microfluidic devices offer a unique technological approach to build tumor-on-a-chip reproducing the multiple layers of complexity of cancer-immune system crosstalk. Here, we seek to review the most important biological and engineering developments of microfluidic platforms for studying cancer-immune system interactions, in both solid and hematological tumors, highlighting the role of the vascular component in immune trafficking. Emphasis is given to image processing and related algorithms for real-time monitoring and quantitative evaluation of the cellular response to microenvironmental dynamic changes. The described approaches represent a valuable tool for preclinical evaluation of immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefania Parlato
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy.
| | | | | | | | | | | |
Collapse
|