1
|
Hammel JH, Arneja A, Cunningham J, Wang M, Schumaecker S, Orihuela YM, Ozulumba T, Zatorski JM, Braciale TJ, Luckey CJ, Pompano RR, Munson JM. Interstitial fluid flow in an engineered human lymph node stroma model modulates T cell egress and stromal change. APL Bioeng 2025; 9:026105. [PMID: 40191604 PMCID: PMC11972091 DOI: 10.1063/5.0247363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/15/2025] [Indexed: 04/09/2025] Open
Abstract
The lymph node (LN) performs essential roles in immunosurveillance throughout the body. Developing in vitro models of this key tissue is of great importance to enhancing physiological relevance in immunoengineering. The LN consists of stromal populations and immune cells, which are highly organized and bathed in constant interstitial fluid flow (IFF). The stroma, notably the fibroblastic reticular cells (FRCs) and the lymphatic endothelial cells (LECs), play crucial roles in guiding T cell migration and are known to be sensitive to fluid flow. During inflammation, interstitial fluid flow rates drastically increase in the LN. It is unknown how these altered flow rates impact crosstalk and cell behavior in the LN, and most existing in vitro models focus on the interactions between T cells, B cells, and dendritic cells rather than with the stroma. To address this gap, we developed a human engineered model of the LN stroma consisting of FRC-laden hydrogel above a monolayer of LECs in a tissue culture insert with gravity-driven interstitial flow. We found that FRCs had enhanced coverage and proliferation in response to high flow rates, while LECs experienced decreased barrier integrity. We added CD4+ and CD8+ T cells and found that their egress was significantly decreased in the presence of interstitial flow, regardless of magnitude. Interestingly, 3.0 μm/s flow, but not 0.8 μm/s flow, correlated with enhanced inflammatory cytokine secretion in the LN stroma. Overall, we demonstrate that interstitial flow is an essential consideration in the lymph node for modulating LN stroma morphology, T cell migration, and inflammation.
Collapse
Affiliation(s)
| | - Abhinav Arneja
- University of Virginia Department of Pathology, 415 Lane Road, Charlottesville, Virginia 22908, USA
| | - Jessica Cunningham
- Virginia Tech Fralin Biomedical Research Institute, 4 Riverside Circle, Roanoke, Virginia 24016, USA
| | - Maosen Wang
- Virginia Tech Fralin Biomedical Research Institute, 4 Riverside Circle, Roanoke, Virginia 24016, USA
| | - Sophia Schumaecker
- Virginia Tech Fralin Biomedical Research Institute, 4 Riverside Circle, Roanoke, Virginia 24016, USA
| | | | - Tochukwu Ozulumba
- University of Virginia Department of Chemistry, 409 McCormick Road, Charlottesville, Virginia 22904, USA
| | - Jonathan M. Zatorski
- University of Virginia Department of Chemistry, 409 McCormick Road, Charlottesville, Virginia 22904, USA
| | - Thomas J. Braciale
- University of Virginia Department of Pathology, 415 Lane Road, Charlottesville, Virginia 22908, USA
| | - Chance John Luckey
- University of Virginia Department of Pathology, 415 Lane Road, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
2
|
Polacheck WJ, Dixon JB, Aw WY. Understanding the Lymphatic System: Tissue-on-Chip Modeling. Annu Rev Biomed Eng 2025; 27:73-100. [PMID: 39841937 DOI: 10.1146/annurev-bioeng-110222-100246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The lymphatic vasculature plays critical roles in maintaining fluid homeostasis, transporting lipid, and facilitating immune surveillance. A growing body of work has identified lymphatic dysfunction as contributing to the severity of myriad diseases and to systemic inflammation, as well as modulating drug responses. Here, we review efforts to reconstruct lymphatic vessels in vitro toward establishing humanized, functional models to advance understanding of lymphatic biology and pathophysiology. We first review lymphatic endothelial cell biology and the biophysical lymphatic microenvironment, with a focus on features that are unique to the lymphatics and that have been used as design parameters for lymphatic-on-chip devices. We then discuss the state of the art for recapitulating lymphatic function in vitro, and we acknowledge limitations and challenges to current approaches. Finally, we discuss opportunities and the need for further development of microphysiological lymphatic systems to bridge the gap in model systems between lymphatic cell culture and animal physiology.
Collapse
Affiliation(s)
- William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina, USA;
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J Brandon Dixon
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Wen Yih Aw
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina, USA;
| |
Collapse
|
3
|
Morgaenko K, Arneja A, Ball AG, Putelo AM, Munson JM, Rutkowski MR, Pompano RR. Ex Vivo Model of Breast Cancer Cell Invasion in Live Lymph Node Tissue. ACS Pharmacol Transl Sci 2025; 8:690-705. [PMID: 40109746 PMCID: PMC11915036 DOI: 10.1021/acsptsci.4c00431] [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: 07/19/2024] [Revised: 12/12/2024] [Accepted: 01/27/2025] [Indexed: 03/22/2025]
Abstract
Lymph nodes (LNs) are common sites of metastatic invasion in breast cancer, often preceding spread to distant organs and serving as key indicators of clinical disease progression. However, the mechanisms of cancer cell invasion into LNs are not well understood. Existing in vivo models struggle to isolate the specific impacts of the tumor-draining lymph node (TDLN) milieu on cancer cell invasion due to the coevolving relationship between TDLNs and the upstream tumor. To address these limitations, we used live ex vivo LN tissue slices with intact chemotactic function to model cancer cell spread within a spatially organized microenvironment. After showing that BRPKp110 breast cancer cells were chemoattracted to factors secreted by naïve LN tissue in a 3D migration assay, we demonstrated that ex vivo LN slices could support cancer cell seeding, invasion, and spread. This novel approach revealed dynamic, preferential cancer cell invasion within specific anatomical regions of LNs, particularly the subcapsular sinus (SCS) and cortex, as well as chemokine-rich domains of immobilized CXCL13 and CCL1. While CXCR5 was necessary for a portion of BRPKp110 invasion into naïve LNs, disruption of CXCR5/CXCL13 signaling alone was insufficient to prevent invasion toward CXCL13-rich domains. Finally, we extended this system to premetastatic TDLNs, where the ex vivo model predicted a lower invasion of cancer cells that was not due to diminished chemokine secretion. In summary, this innovative ex vivo model of cancer cell spread in live LN slices provides a platform to investigate cancer invasion within the intricate tissue microenvironment, supporting time-course analysis and parallel read-outs. We anticipate that this system will enable further research into cancer-immune interactions and allow for isolation of specific factors that make TDLNs resistant to cancer cell invasion, which is challenging to dissect in vivo.
Collapse
Affiliation(s)
- Katerina Morgaenko
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
- Carter
Immunology Center and University of Virginia Cancer Center, University
of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
| | - Abhinav Arneja
- Department
of Pathology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Alexander G. Ball
- Carter
Immunology Center and University of Virginia Cancer Center, University
of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
- Department
of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Audrey M. Putelo
- Department
of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Jennifer M. Munson
- Department
of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, Virginia 24016, United States
| | - Melanie R. Rutkowski
- Department
of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Rebecca R. Pompano
- Department
of Biomedical Engineering, University of
Virginia, Charlottesville, Virginia 22904, United States
- Carter
Immunology Center and University of Virginia Cancer Center, University
of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
- Department
of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
4
|
Mazzaglia C, Shery Huang YY, Shields JD. Advancing tumor microenvironment and lymphoid tissue research through 3D bioprinting and biofabrication. Adv Drug Deliv Rev 2025; 217:115485. [PMID: 39653084 DOI: 10.1016/j.addr.2024.115485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Cancer progression is significantly influenced by the complex interactions within the tumor microenvironment (TME). Immune cells, in particular, play a critical role by infiltrating tumors from the circulation and surrounding lymphoid tissues in an attempt to control their spread. However, they often fail in this task. Current in vivo and in vitro preclinical models struggle to fully capture these intricate interactions affecting our ability to understand immune evasion and predict drugs behaviour in the clinic. To address this challenge, biofabrication and particularly 3D bioprinting has emerged as a promising tool for modeling both tumors and the immune system. Its ability to incorporate multiple cell types into 3D matrices, enable tissue compartmentalization with high spatial accuracy, and integrate vasculature makes it a valuable approach. Nevertheless, limited research has focused on capturing the complex tumor-immune interplay in vitro. This review highlights the composition and significance of the TME, the architecture and function of lymphoid tissues, and innovative approaches to modeling their interactions in vitro, while proposing the concept of an extended TME.
Collapse
Affiliation(s)
- Corrado Mazzaglia
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Center for Life Nano, and Neuro-Science of Istituto Italiano di Tecnologia (IIT), Rome 00161, Italy.
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland
| | - Jacqueline D Shields
- Translational Medical Sciences, School of Medicine, University of Nottingham, Biodiscovery Institute, Nottingham, the United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
5
|
Janssen R, Benito-Zarza L, Cleijpool P, Valverde MG, Mihăilă SM, Bastiaan-Net S, Garssen J, Willemsen LEM, Masereeuw R. Biofabrication Directions in Recapitulating the Immune System-on-a-Chip. Adv Healthc Mater 2025; 14:e2304569. [PMID: 38625078 DOI: 10.1002/adhm.202304569] [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: 12/21/2023] [Revised: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Ever since the implementation of microfluidics in the biomedical field, in vitro models have experienced unprecedented progress that has led to a new generation of highly complex miniaturized cell culture platforms, known as Organs-on-a-Chip (OoC). These devices aim to emulate biologically relevant environments, encompassing perfusion and other mechanical and/or biochemical stimuli, to recapitulate key physiological events. While OoCs excel in simulating diverse organ functions, the integration of the immune organs and immune cells, though recent and challenging, is pivotal for a more comprehensive representation of human physiology. This comprehensive review covers the state of the art in the intricate landscape of immune OoC models, shedding light on the pivotal role of biofabrication technologies in bridging the gap between conceptual design and physiological relevance. The multifaceted aspects of immune cell behavior, crosstalk, and immune responses that are aimed to be replicated within microfluidic environments, emphasizing the need for precise biomimicry are explored. Furthermore, the latest breakthroughs and challenges of biofabrication technologies in immune OoC platforms are described, guiding researchers toward a deeper understanding of immune physiology and the development of more accurate and human predictive models for a.o., immune-related disorders, immune development, immune programming, and immune regulation.
Collapse
Affiliation(s)
- Robine Janssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Laura Benito-Zarza
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Pim Cleijpool
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Marta G Valverde
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Silvia M Mihăilă
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, 6708 WG, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Danone Global Research & Innovation Center, Danone Nutricia Research B.V., Utrecht, 3584 CT, The Netherlands
| | - Linette E M Willemsen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| |
Collapse
|
6
|
Zatorski JM, Raskovic D, Arneja A, Kiridena S, Ozulumba T, Hammel JH, Anbaei P, Ortiz-Cárdenas JE, Braciale TJ, Munson JM, Luckey CJ, Pompano RR. Initiation of primary T cell-B cell interactions and extrafollicular antibody responses in an organized microphysiological model of the human lymph node. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632545. [PMID: 39868310 PMCID: PMC11761657 DOI: 10.1101/2025.01.12.632545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Antibody production is central to protection against new pathogens and cancers, as well as to certain forms of autoimmunity. Antibodies often originate in the lymph node (LN), specifically at the extrafollicular border of B cell follicles, where T and B lymphocytes physically interact to drive B cell maturation into antibody-secreting plasmablasts. In vitro models of this process are sorely needed to predict aspects of the human immune response. Microphysiological systems (MPSs) offer the opportunity to approximate the lymphoid environment, but so far have focused primarily on memory recall responses to antigens previously encountered by donor cells. To date, no 3D culture system has replicated the engagement between T cells and B cells (T-B interaction) that leads to antibody production when starting with naïve cells. Here, we developed a LN-MPS to model early T-B interactions at the extrafollicular border built from primary, naïve human lymphocytes encapsulated within a collagen-based 3D matrix. Within the MPS, naïve T cells exhibited CCL21-dependent chemotaxis and chemokinesis as predicted. Naïve T and B cells were successfully skewed on chip to an early T follicular helper (pre-Tfh) and activated state, respectively, and co-culture of the latter cells led to CD38+ plasmablast cells and T cell dependent production of IgM. These responses required differentiation of the T cells into pre-Tfhs, physical cell-cell contact, and were sensitive to the ratio at which pre-Tfh and activated B cells were seeded on-chip. Dependence on T cell engagement was greatest at a 1:5 T:B ratio, while cell proliferation and CD38+ signal was greatest at a 1:1 T:B ratio. Furthermore, plasmablast formation was established starting from naïve T and B cells on-chip. We envision that this MPS model of primary lymphocyte physiology will enable new mechanistic analyses of human humoral immunity in vitro.
Collapse
Affiliation(s)
- Jonathan M Zatorski
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Djuro Raskovic
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Saweetha Kiridena
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Tochukwu Ozulumba
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Jennifer H Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Roanoke, VA
| | - Parastoo Anbaei
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Jennifer E Ortiz-Cárdenas
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
- Stanford University, Department of Bioengineering, 443 Via Ortega, Rm 119, Stanford, CA 94305, United States
| | - Thomas J Braciale
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Jennifer M Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Roanoke, VA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Rebecca R Pompano
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Sciences, Thornton Hall, 351 McCormick Rd, Charlottesville, VA 22904
| |
Collapse
|
7
|
Hammel JH, Arneja A, Cunningham J, Wang M, Schumaecker S, Orihuela YM, Ozulumba T, Zatorski J, Braciale TJ, Luckey CJ, Pompano RR, Munson JM. Engineered human lymph node stroma model for examining interstitial fluid flow and T cell egress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.622729. [PMID: 39677702 PMCID: PMC11642859 DOI: 10.1101/2024.12.03.622729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The lymph node (LN) performs essential roles in immunosurveillance throughout the body. Developing in vitro models of this key tissue is of great importance to enhancing physiological relevance in immunoengineering. The LN consists of stromal populations and immune cells, which are highly organized and bathed in constant interstitial flow. The stroma, notably the fibroblastic reticular cells (FRCs) and the lymphatic endothelial cells (LECs), play crucial roles in guiding T cell migration and are known to be sensitive to fluid flow. During inflammation, interstitial fluid flow rates drastically increase in the LN. It is unknown how these altered flow rates impact crosstalk and cell behavior in the LN, and most existing in vitro models focus on the interactions between T cells, B cells, and dendritic cells rather than with the stroma. To address this gap, we developed a human engineered model of the LN stroma consisting of FRC-laden hydrogel above a monolayer of LECs in a tissue culture insert with gravity-driven interstitial flow. We found that FRCs had enhanced coverage and proliferation in response to high flow rates, while LECs experienced decreased barrier integrity. We added CD4+ and CD8+ T cells and found that their egress was significantly decreased in the presence of interstitial flow, regardless of magnitude. Interestingly, 3.0 µm/s flow, but not 0.8 µm/s flow, correlated with enhanced inflammatory cytokine secretion in the LN stroma. Overall, we demonstrate that interstitial flow is an essential consideration in the lymph node for modulating LN stroma morphology, T cell migration, and inflammation.
Collapse
|
8
|
Girelli A, Giantesio G, Musesti A, Penta R. Multiscale computational analysis of the steady fluid flow through a lymph node. Biomech Model Mechanobiol 2024; 23:2005-2023. [PMID: 39320689 PMCID: PMC11554713 DOI: 10.1007/s10237-024-01879-7] [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: 02/29/2024] [Accepted: 07/12/2024] [Indexed: 09/26/2024]
Abstract
Lymph Nodes (LNs) are crucial to the immune and lymphatic systems, filtering harmful substances and regulating lymph transport. LNs consist of a lymphoid compartment (LC) that forms a porous bulk region, and a subcapsular sinus (SCS), which is a free-fluid region. Mathematical and mechanical challenges arise in understanding lymph flow dynamics. The highly vascularized lymph node connects the lymphatic and blood systems, emphasizing its essential role in maintaining the fluid balance in the body. In this work, we describe a mathematical model in a steady setting to describe the lymph transport in a lymph node. We couple the fluid flow in the SCS governed by an incompressible Stokes equation with the fluid flow in LC, described by a model obtained by means of asymptotic homogenisation technique, taking into account the multiscale nature of the node and the fluid exchange with the blood vessels inside it. We solve this model using numerical simulations and we analyze the lymph transport inside the node to elucidate its regulatory mechanisms and significance. Our results highlight the crucial role of the microstructure of the lymph node in regularising its fluid balance. These results can pave the way to a better understanding of the mechanisms underlying the lymph node's multiscale functionalities which can be significantly affected by specific physiological and pathological conditions, such as those characterising malignant tissues.
Collapse
Affiliation(s)
- Alberto Girelli
- Dipartimento di Matematica e Fisica "N. Tartaglia", Università Cattolica del Sacro Cuore, Brescia, Italy
| | - Giulia Giantesio
- Dipartimento di Matematica e Fisica "N. Tartaglia", Università Cattolica del Sacro Cuore, Brescia, Italy
- Mathematics for Technology, Medicine and Biosciences, Università degli Studi di Ferrara, Ferrara, Italy
| | - Alessandro Musesti
- Dipartimento di Matematica e Fisica "N. Tartaglia", Università Cattolica del Sacro Cuore, Brescia, Italy
| | - Raimondo Penta
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK.
| |
Collapse
|
9
|
Neagu AN, Whitham D, Bruno P, Versaci N, Biggers P, Darie CC. Tumor-on-chip platforms for breast cancer continuum concept modeling. Front Bioeng Biotechnol 2024; 12:1436393. [PMID: 39416279 PMCID: PMC11480020 DOI: 10.3389/fbioe.2024.1436393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Our previous article entitled "Proteomics and its applications in breast cancer", proposed a Breast Cancer Continuum Concept (BCCC), including a Breast Cancer Cell Continuum Concept as well as a Breast Cancer Proteomic Continuum Concept. Breast cancer-on-chip (BCoC), breast cancer liquid biopsy-on-chip (BCLBoC), and breast cancer metastasis-on-chip (BCMoC) models successfully recapitulate and reproduce in vitro the principal mechanisms and events involved in BCCC. Thus, BCoC, BCLBoC, and BCMoC platforms allow for multiple cell lines co-cultivation to reproduce BC hallmark features, recapitulating cell proliferation, cell-to-cell communication, BC cell-stromal crosstalk and stromal activation, effects of local microenvironmental conditions on BC progression, invasion/epithelial-mesenchymal transition (EMT)/migration, intravasation, dissemination through blood and lymphatic circulation, extravasation, distant tissues colonization, and immune escape of cancer cells. Moreover, tumor-on-chip platforms are used for studying the efficacy and toxicity of chemotherapeutic drugs/nano-drugs or nutraceuticals. Therefore, the aim of this review is to summarize and analyse the main bio-medical roles of on-chip platforms that can be used as powerful tools to study the metastatic cascade in BC. As future direction, integration of tumor-on-chip platforms and proteomics-based specific approaches can offer important cues about molecular profile of the metastatic cascade, alowing for novel biomarker discovery. Novel microfluidics-based platforms integrating specific proteomic landscape of human milk, urine, and saliva could be useful for early and non-invasive BC detection. Also, risk-on-chip models may improve BC risk assessment and prevention based on the identification of biomarkers of risk. Moreover, multi-organ-on-chip systems integrating patient-derived BC cells and patient-derived scaffolds have a great potential to study BC at integrative level, due to the systemic nature of BC, for personalized and precision medicine. We also emphasized the strengths and weaknesses of BCoC and BCMoC platforms.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Nicholas Versaci
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Peter Biggers
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY, United States
| |
Collapse
|
10
|
Girelli A. A quasilinear hyperbolic one-dimensional model of the lymph flow through a lymphangion with valve dynamics and a contractile wall. Comput Methods Biomech Biomed Engin 2024:1-16. [PMID: 39262168 DOI: 10.1080/10255842.2024.2399769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/03/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
This paper presents a one-dimensional model that describes fluid flow in lymphangions, the segments of lymphatic vessels between valves, using quasilinear hyperbolic systems. The model incorporates a phenomenological pressure-cross-sectional area relationship based on existing literature. Numerical solutions of the differential equations align with known results, offering insights into lymphatic flow dynamics. This model enhances the understanding of lymph movement through the lymphatic system, driven by lymphangion contractions.
Collapse
Affiliation(s)
- Alberto Girelli
- Dipartimento di Matematica e Fisica "N. Tartaglia", Università Cattolica del Sacro Cuore, Brescia, Italy
| |
Collapse
|
11
|
Gopallawa I, Gupta C, Jawa R, Cyril A, Jawa V, Chirmule N, Gujar V. Applications of Organoids in Advancing Drug Discovery and Development. J Pharm Sci 2024; 113:2659-2667. [PMID: 39002723 DOI: 10.1016/j.xphs.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
Organoids are small, self-organizing three-dimensional cell cultures that are derived from stem cells or primary organs. These cultures replicate the complexity of an organ, which cannot be achieved by single-cell culture systems. Organoids can be used in testing of new drugs instead of animals. Development and validation of organoids is thus important to reduce the reliance on animals for drug testing. In this review, we have discussed the developmental and regulatory aspects of organoids and highlighted their importance in drug development. We have first summarized different types of culture-based organoid systems such as submerged Matrigel, micro-fluidic 3D cultures, inducible pluripotent stem cells, and air-liquid interface cultures. These systems help us understand the intricate interplay between cells and their surrounding milieu for identifying functions of target receptors, soluble factors, and spatial interactions. Further, we have discussed the advances in humanized severe-combined immunodeficiency mouse models and their applications in the pharmacology of immune-oncology. Since regulatory aspects are important in using organoids for drug development, we have summarized FDA and EMA regulations on organoid research to support pre-clinical studies. Finally, we have included some unique studies highlighting the use of organoids in studying infectious diseases, cancer, and fundamental biology. These studies also exemplify the latest technological advances in organoid development resulting in improved efficiency. Overall, this review comprehensively summarizes the applications of organoids in early drug development during discovery and pre-clinical studies.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | | | - Rayan Jawa
- University of Pennsylvania, Philadelphia, PA, USA
| | - Arya Cyril
- Department of Psychology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Vibha Jawa
- Bristol Myers Squibb, Lawrenceville, NY, USA.
| | | | - Vikramsingh Gujar
- Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| |
Collapse
|
12
|
Morgaenko K, Arneja A, Ball AG, Putelo AM, Munson JM, Rutkowski MR, Pompano RR. Ex vivo model of breast cancer cell invasion in live lymph node tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.601753. [PMID: 39091774 PMCID: PMC11291011 DOI: 10.1101/2024.07.18.601753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymph nodes (LNs) are common sites of metastatic invasion in breast cancer, often preceding spread to distant organs and serving as key indicators of clinical disease progression. However, the mechanisms of cancer cell invasion into LNs are not well understood. Existing in vivo models struggle to isolate the specific impacts of the tumor-draining lymph node (TDLN) milieu on cancer cell invasion due to the co-evolving relationship between TDLNs and the upstream tumor. To address these limitations, we used live ex vivo LN tissue slices with intact chemotactic function to model cancer cell spread within a spatially organized microenvironment. After showing that BRPKp110 breast cancer cells were chemoattracted to factors secreted by naïve LN tissue in a 3D migration assay, we demonstrated that ex vivo LN slices could support cancer cell seeding, invasion, and spread. This novel approach revealed dynamic, preferential cancer cell invasion within specific anatomical regions of LNs, particularly the subcapsular sinus (SCS) and cortex, as well as chemokine-rich domains of immobilized CXCL13 and CCL1. While CXCR5 was necessary for a portion of BRPKp110 invasion into naïve LNs, disruption of CXCR5/CXCL13 signaling alone was insufficient to prevent invasion towards CXCL13-rich domains. Finally, we extended this system to pre-metastatic TDLNs, where the ex vivo model predicted a lower invasion of cancer cells. The reduced invasion was not due to diminished chemokine secretion, but it correlated with elevated intranodal IL-21. In summary, this innovative ex vivo model of cancer cell spread in live LN slices provides a platform to investigate cancer invasion within the intricate tissue microenvironment, supporting time-course analysis and parallel read-outs. We anticipate that this system will enable further research into cancer-immune interactions and allow isolation of specific factors that make TDLNs resistant to cancer cell invasion, which are challenging to dissect in vivo.
Collapse
Affiliation(s)
- Katerina Morgaenko
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Alexander G Ball
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Audrey M Putelo
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Jennifer M Munson
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA, United States
| | - Melanie R Rutkowski
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Rebecca R Pompano
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
13
|
Mazzaglia C, Munir H, Lei IM, Gerigk M, Huang YYS, Shields JD. Modeling Structural Elements and Functional Responses to Lymphatic-Delivered Cues in a Murine Lymph Node on a Chip. Adv Healthc Mater 2024; 13:e2303720. [PMID: 38626388 DOI: 10.1002/adhm.202303720] [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: 10/26/2023] [Revised: 04/08/2024] [Indexed: 04/18/2024]
Abstract
Lymph nodes (LNs) are organs of the immune system, critical for maintenance of homeostasis and initiation of immune responses, yet there are few models that accurately recapitulate LN functions in vitro. To tackle this issue, an engineered murine LN (eLN) has been developed, replicating key cellular components of the mouse LN; incorporating primary murine lymphocytes, fibroblastic reticular cells, and lymphatic endothelial cells. T and B cell compartments are incorporated within the eLN that mimic LN cortex and paracortex architectures. When challenged, the eLN elicits both robust inflammatory responses and antigen-specific immune activation, showing that the system can differentiate between non specific and antigen-specific stimulation and can be monitored in real time. Beyond immune responses, this model also enables interrogation of changes in stromal cells, thus permitting investigations of all LN cellular components in homeostasis and different disease settings, such as cancer. Here, how LN behavior can be influenced by murine melanoma-derived factors is presented. In conclusion, the eLN model presents a promising platform for in vitro study of LN biology that will enhance understanding of stromal and immune responses in the murine LN, and in doing so will enable development of novel therapeutic strategies to improve LN responses in disease.
Collapse
Affiliation(s)
- Corrado Mazzaglia
- MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, UK
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Hafsa Munir
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz), 55131, Mainz, Germany
- Division of Dermal Oncoimmunology, German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
| | - Iek Man Lei
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ, UK
| | - Magda Gerigk
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ, UK
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ, UK
| | - Jacqueline D Shields
- MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, UK
- Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| |
Collapse
|
14
|
Girelli A, Giantesio G, Musesti A, Penta R. Multiscale homogenization for dual porosity time-dependent Darcy-Brinkman/Darcy coupling and its application to the lymph node. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231983. [PMID: 39021765 PMCID: PMC11253036 DOI: 10.1098/rsos.231983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/17/2024] [Indexed: 07/20/2024]
Abstract
We study the coupling between time-dependent Darcy-Brinkman and the Darcy equations at the microscale subjected to inhomogeneous body forces and initial conditions to describe a double porosity problem. We derive the homogenized governing equations for this problem using the asymptotic homogenization technique, and as macroscopic results, we obtain a coupling between two Darcy equations, one of which with memory effects, with mass exchange between phases. The memory effects are a consequence of considering the time dependence in the Darcy-Brinkman equation, and they allow us to study in more detail the role of time in the problem under consideration. After the formulation of the model, we solve it in a simplified setting and we use it to describe the movement of fluid within a vascularized lymph node.
Collapse
Affiliation(s)
- A. Girelli
- Dipartimento di Matematica e Fisica ‘N. Tartaglia’, Università Cattolica del Sacro Cuore, Brescia, Italy
| | - G. Giantesio
- Dipartimento di Matematica e Fisica ‘N. Tartaglia’, Università Cattolica del Sacro Cuore, Brescia, Italy
- ‘Mathematics for Technology, Medicine and Biosciences’, Università degli Studi di Ferrara, Ferrara, Italy
| | - A. Musesti
- Dipartimento di Matematica e Fisica ‘N. Tartaglia’, Università Cattolica del Sacro Cuore, Brescia, Italy
| | - R. Penta
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| |
Collapse
|
15
|
Liang L, Song X, Zhao H, Lim CT. Insights into the mechanobiology of cancer metastasis via microfluidic technologies. APL Bioeng 2024; 8:021506. [PMID: 38841688 PMCID: PMC11151435 DOI: 10.1063/5.0195389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
During cancer metastasis, cancer cells will encounter various microenvironments with diverse physical characteristics. Changes in these physical characteristics such as tension, stiffness, viscosity, compression, and fluid shear can generate biomechanical cues that affect cancer cells, dynamically influencing numerous pathophysiological mechanisms. For example, a dense extracellular matrix drives cancer cells to reorganize their cytoskeleton structures, facilitating confined migration, while this dense and restricted space also acts as a physical barrier that potentially results in nuclear rupture. Identifying these pathophysiological processes and understanding their underlying mechanobiological mechanisms can aid in the development of more effective therapeutics targeted to cancer metastasis. In this review, we outline the advances of engineering microfluidic devices in vitro and their role in replicating tumor microenvironment to mimic in vivo settings. We highlight the potential cellular mechanisms that mediate their ability to adapt to different microenvironments. Meanwhile, we also discuss some important mechanical cues that still remain challenging to replicate in current microfluidic devices in future direction. While much remains to be explored about cancer mechanobiology, we believe the developments of microfluidic devices will reveal how these physical cues impact the behaviors of cancer cells. It will be crucial in the understanding of cancer metastasis, and potentially contributing to better drug development and cancer therapy.
Collapse
Affiliation(s)
- Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xiao Song
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | | |
Collapse
|
16
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
17
|
Wang Q, Yang Y, Chen Z, Li B, Niu Y, Li X. Lymph Node-on-Chip Technology: Cutting-Edge Advances in Immune Microenvironment Simulation. Pharmaceutics 2024; 16:666. [PMID: 38794327 PMCID: PMC11124897 DOI: 10.3390/pharmaceutics16050666] [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: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-a-chip technology is attracting growing interest across various domains as a crucial platform for drug screening and testing and is set to play a significant role in precision medicine research. Lymph nodes, being intricately structured organs essential for the body's adaptive immune responses to antigens and foreign particles, are pivotal in assessing the immunotoxicity of novel pharmaceuticals. Significant progress has been made in research on the structure and function of the lymphatic system. However, there is still an urgent need to develop prospective tools and techniques to delve deeper into its role in various diseases' pathological and physiological processes and to develop corresponding immunotherapeutic therapies. Organ chips can accurately reproduce the specific functional areas in lymph nodes to better simulate the complex microstructure of lymph nodes and the interactions between different immune cells, which is convenient for studying specific biological processes. This paper reviews existing lymph node chips and their design approaches. It discusses the applications of the above systems in modeling immune cell motility, cell-cell interactions, vaccine responses, drug testing, and cancer research. Finally, we summarize the challenges that current research faces in terms of structure, cell source, and extracellular matrix simulation of lymph nodes, and we provide an outlook on the future direction of integrated immune system chips.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoqiong Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Q.W.); (Y.Y.); (Z.C.); (B.L.); (Y.N.)
| |
Collapse
|
18
|
Camargo CP, Alapan Y, Muhuri AK, Lucas SN, Thomas SN. Single-cell adhesive profiling in an optofluidic device elucidates CD8 + T lymphocyte phenotypes in inflamed vasculature-like microenvironments. CELL REPORTS METHODS 2024; 4:100743. [PMID: 38554703 PMCID: PMC11046032 DOI: 10.1016/j.crmeth.2024.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/28/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
Tissue infiltration by circulating leukocytes occurs via adhesive interactions with the local vasculature, but how the adhesive quality of circulating cells guides the homing of specific phenotypes to different vascular microenvironments remains undefined. We developed an optofluidic system enabling fluorescent labeling of photoactivatable cells based on their adhesive rolling velocity in an inflamed vasculature-mimicking microfluidic device under physiological fluid flow. In so doing, single-cell level multidimensional profiling of cellular characteristics could be characterized and related to the associated adhesive phenotype. When applied to CD8+ T cells, ligand/receptor expression profiles and subtypes associated with adhesion were revealed, providing insight into inflamed tissue infiltration capabilities of specific CD8+ T lymphocyte subsets and how local vascular microenvironmental features may regulate the quality of cellular infiltration. This methodology facilitates rapid screening of cell populations for enhanced homing capabilities under defined biochemical and biophysical microenvironments, relevant to leukocyte homing modulation in multiple pathologies.
Collapse
Affiliation(s)
- Camila P Camargo
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Yunus Alapan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Abir K Muhuri
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA
| | - Samuel N Lucas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta 30332, GA, USA
| | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta 30332, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta 30332, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta 30332, GA, USA; Winship Cancer Institute, Emory University, Atlanta 30322, GA, USA.
| |
Collapse
|
19
|
da Costa Sousa MG, Vignolo SM, Franca CM, Mereness J, Alves Fraga MA, Silva-Sousa AC, Benoit DSW, Bertassoni LE. Engineering models of head and neck and oral cancers on-a-chip. BIOMICROFLUIDICS 2024; 18:021502. [PMID: 38464668 PMCID: PMC10919958 DOI: 10.1063/5.0186722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
Head and neck cancers (HNCs) rank as the sixth most common cancer globally and result in over 450 000 deaths annually. Despite considerable advancements in diagnostics and treatment, the 5-year survival rate for most types of HNCs remains below 50%. Poor prognoses are often attributed to tumor heterogeneity, drug resistance, and immunosuppression. These characteristics are difficult to replicate using in vitro or in vivo models, culminating in few effective approaches for early detection and therapeutic drug development. Organs-on-a-chip offer a promising avenue for studying HNCs, serving as microphysiological models that closely recapitulate the complexities of biological tissues within highly controllable microfluidic platforms. Such systems have gained interest as advanced experimental tools to investigate human pathophysiology and assess therapeutic efficacy, providing a deeper understanding of cancer pathophysiology. This review outlines current challenges and opportunities in replicating HNCs within microphysiological systems, focusing on mimicking the soft, glandular, and hard tissues of the head and neck. We further delve into the major applications of organ-on-a-chip models for HNCs, including fundamental research, drug discovery, translational approaches, and personalized medicine. This review emphasizes the integration of organs-on-a-chip into the repertoire of biological model systems available to researchers. This integration enables the exploration of unique aspects of HNCs, thereby accelerating discoveries with the potential to improve outcomes for HNC patients.
Collapse
Affiliation(s)
| | | | | | - Jared Mereness
- Departments of Biomedical Engineering and Dermatology and Center for Musculoskeletal Research, University of Rochester, 601 Elmwood Ave, Rochester, New York 14642, USA
| | | | - Alice Corrêa Silva-Sousa
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo. Av. do Café - Subsetor Oeste—11 (N-11), Ribeirão Preto, SP, 14040-904, Brazil
| | | | | |
Collapse
|
20
|
Janssen R, de Kleer JWM, Heming B, Bastiaan-Net S, Garssen J, Willemsen LEM, Masereeuw R. Food allergen sensitization on a chip: the gut-immune-skin axis. Trends Biotechnol 2024; 42:119-134. [PMID: 37580191 DOI: 10.1016/j.tibtech.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023]
Abstract
The global population is growing, rapidly increasing the demand for sustainable, novel, and safe food proteins with minimal risks of food allergy. In vitro testing of allergy-sensitizing capacity is predominantly based on 2D assays. However, these lack the 3D environment and crosstalk between the gut, skin, and immune cells essential for allergy prediction. Organ-on-a-chip (OoC) technologies are promising to study type 2 immune activation required for sensitization, initiated in the small intestine or skin, in interlinked systems. Increasing the mechanistic understanding and, moreover, finding new strategies to study interorgan communication is of importance to recapitulate food allergen sensitization in vitro. Here, we outline recently developed OoC platforms and discuss the features needed for reliable prediction of sensitizing allergenicity of proteins.
Collapse
Affiliation(s)
- Robine Janssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Janna W M de Kleer
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Bo Heming
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food and Biobased Research, Wageningen University and Research, Wageningen, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands; Danone Nutricia Research B.V., Utrecht, The Netherlands
| | - Linette E M Willemsen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
21
|
Chen R, Chen L, Wang C, Zhu H, Gu L, Li Y, Xiong X, Chen G, Jian Z. CAR-T treatment for cancer: prospects and challenges. Front Oncol 2023; 13:1288383. [PMID: 38115906 PMCID: PMC10728652 DOI: 10.3389/fonc.2023.1288383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Chimeric antigen receptor (CAR-T) cell therapy has been widely used in hematological malignancies and has achieved remarkable results, but its long-term efficacy in solid tumors is greatly limited by factors such as the tumor microenvironment (TME). In this paper, we discuss the latest research and future views on CAR-T cell cancer immunotherapy, compare the different characteristics of traditional immunotherapy and CAR-T cell therapy, introduce the latest progress in CAR-T cell immunotherapy, and analyze the obstacles that hinder the efficacy of CAR-T cell therapy, including immunosuppressive factors, metabolic energy deficiency, and physical barriers. We then further discuss the latest therapeutic strategies to overcome these barriers, as well as management decisions regarding the possible safety issues of CAR-T cell therapy, to facilitate solutions to the limited use of CAR-T immunotherapy.
Collapse
Affiliation(s)
- Ran Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chaoqun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuntao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gang Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Kraus S, Lee E. A human initial lymphatic chip reveals distinct mechanisms of primary lymphatic valve dysfunction in acute and chronic inflammation. LAB ON A CHIP 2023; 23:5180-5194. [PMID: 37981867 PMCID: PMC10908576 DOI: 10.1039/d3lc00486d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Interstitial fluid uptake and retention by lymphatic vessels (LVs) play a role in maintaining interstitial fluid homeostasis. While it is well-established that intraluminal lymphatic valves in the collecting LVs prevent fluid backflow (secondary lymphatic valves), a separate valve system in the initial LVs that only permits interstitial fluid influx into the LVs, preventing fluid leakage back to the interstitium (primary lymphatic valves), remains incompletely understood. Although lymphatic dysfunction is commonly observed in inflammation and autoimmune diseases, how the primary lymphatic valves are affected by acute and chronic inflammation has scarcely been explored and even less so using in vitro lymphatic models. Here, we developed a human initial lymphatic vessel chip where interstitial fluid pressure and luminal fluid pressure are controlled to examine primary lymph valve function. In normal conditions, lymphatic drainage (fluid uptake) and permeability (fluid leakage) in engineered LVs were maintained high and low, respectively, which was consistent with our understanding of healthy primary lymph valves. Next, we examined the effects of acute and chronic inflammation. Under the acute inflammation condition with a TNF-α treatment (2 hours), degradation of fibrillin and impeded lymphatic drainage were observed, which were reversed by treatment with anti-inflammatory dexamethasone. Surprisingly, the chronic inflammation condition (repeated TNF-α treatments during 48 hours) deposited fibrillin to compensate for the fibrillin loss, showing no change in lymphatic drainage. Instead, the chronic inflammation condition led to cell death and disruption of lymphatic endothelial cell-cell junctions, increasing lymphatic permeability and fluid leakage. Our human lymphatic model shows two distinct mechanisms by which primary lymphatic valve dysfunction occurs in acute and chronic inflammation.
Collapse
Affiliation(s)
- Samantha Kraus
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
23
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
24
|
Kwee BJ, Li X, Nguyen XX, Campagna C, Lam J, Sung KE. Modeling immunity in microphysiological systems. Exp Biol Med (Maywood) 2023; 248:2001-2019. [PMID: 38166397 PMCID: PMC10800123 DOI: 10.1177/15353702231215897] [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] [Indexed: 01/04/2024] Open
Abstract
There is a need for better predictive models of the human immune system to evaluate safety and efficacy of immunomodulatory drugs and biologics for successful product development and regulatory approvals. Current in vitro models, which are often tested in two-dimensional (2D) tissue culture polystyrene, and preclinical animal models fail to fully recapitulate the function and physiology of the human immune system. Microphysiological systems (MPSs) that can model key microenvironment cues of the human immune system, as well as of specific organs and tissues, may be able to recapitulate specific features of the in vivo inflammatory response. This minireview provides an overview of MPS for modeling lymphatic tissues, immunity at tissue interfaces, inflammatory diseases, and the inflammatory tumor microenvironment in vitro and ex vivo. Broadly, these systems have utility in modeling how certain immunotherapies function in vivo, how dysfunctional immune responses can propagate diseases, and how our immune system can combat pathogens.
Collapse
Affiliation(s)
- Brian J Kwee
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19711, USA
| | - Xiaoqing Li
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Xinh-Xinh Nguyen
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Courtney Campagna
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Johnny Lam
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kyung E Sung
- Cellular and Tissue Therapy Branch, Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
25
|
Lee E, Chan SL, Lee Y, Polacheck WJ, Kwak S, Wen A, Nguyen DHT, Kutys ML, Alimperti S, Kolarzyk AM, Kwak TJ, Eyckmans J, Bielenberg DR, Chen H, Chen CS. A 3D biomimetic model of lymphatics reveals cell-cell junction tightening and lymphedema via a cytokine-induced ROCK2/JAM-A complex. Proc Natl Acad Sci U S A 2023; 120:e2308941120. [PMID: 37782785 PMCID: PMC10576061 DOI: 10.1073/pnas.2308941120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/04/2023] [Indexed: 10/04/2023] Open
Abstract
Impaired lymphatic drainage and lymphedema are major morbidities whose mechanisms have remained obscure. To study lymphatic drainage and its impairment, we engineered a microfluidic culture model of lymphatic vessels draining interstitial fluid. This lymphatic drainage-on-chip revealed that inflammatory cytokines that are known to disrupt blood vessel junctions instead tightened lymphatic cell-cell junctions and impeded lymphatic drainage. This opposing response was further demonstrated when inhibition of rho-associated protein kinase (ROCK) was found to normalize fluid drainage under cytokine challenge by simultaneously loosening lymphatic junctions and tightening blood vessel junctions. Studies also revealed a previously undescribed shift in ROCK isoforms in lymphatic endothelial cells, wherein a ROCK2/junctional adhesion molecule-A (JAM-A) complex emerges that is responsible for the cytokine-induced lymphatic junction zippering. To validate these in vitro findings, we further demonstrated in a genetic mouse model that lymphatic-specific knockout of ROCK2 reversed lymphedema in vivo. These studies provide a unique platform to generate interstitial fluid pressure and measure the drainage of interstitial fluid into lymphatics and reveal a previously unappreciated ROCK2-mediated mechanism in regulating lymphatic drainage.
Collapse
Affiliation(s)
- Esak Lee
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Siu-Lung Chan
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Yang Lee
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - William J. Polacheck
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Sukyoung Kwak
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Aiyun Wen
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Duc-Huy T. Nguyen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Matthew L. Kutys
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Stella Alimperti
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Anna M. Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Tae Joon Kwak
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY14853
| | - Jeroen Eyckmans
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| | - Diane R. Bielenberg
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Christopher S. Chen
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA02215
| |
Collapse
|
26
|
Ilan IS, Yslas AR, Peng Y, Lu R, Lee E. A 3D Human Lymphatic Vessel-on-Chip Reveals the Roles of Interstitial Flow and VEGF-A/C for Lymphatic Sprouting and Discontinuous Junction Formation. Cell Mol Bioeng 2023; 16:325-339. [PMID: 37811004 PMCID: PMC10550886 DOI: 10.1007/s12195-023-00780-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/14/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Lymphatic vessels (LVs) maintain fluid homeostasis by draining excess interstitial fluid, which is accomplished by two distinct LVs: initial LVs and collecting LVs. The interstitial fluid is first drained into the initial LVs through permeable "button-like" lymphatic endothelial cell (LEC) junctions. Next, the drained fluid ("lymph") transports to lymph nodes through the collecting LVs with less permeable "zipper-like" junctions that minimize loss of lymph. Despite the significance of LEC junctions in lymphatic drainage and transport, it remains unclear how luminal or interstitial flow affects LEC junctions in vascular endothelial growth factors A and C (VEGF-A and VEGF-C) conditions. Moreover, it remains unclear how these flow and growth factor conditions impact lymphatic sprouting. Methods We developed a 3D human lymphatic vessel-on-chip that can generate four different flow conditions (no flow, luminal flow, interstitial flow, both luminal and interstitial flow) to allow an engineered, rudimentary LV to experience those flows and respond to them in VEGF-A/C. Results We examined LEC junction discontinuities, lymphatic sprouting, LEC junction thicknesses, and cell contractility-dependent vessel diameters in the four different flow conditions in VEGF-A/C. We discovered that interstitial flow in VEGF-C generates discontinuous LEC junctions that may be similar to the button-like junctions with no lymphatic sprouting. However, interstitial flow or both luminal and interstitial flow stimulated lymphatic sprouting in VEGF-A, maintaining zipper-like LEC junctions. LEC junction thickness and cell contractility-dependent vessel diameters were not changed by those conditions. Conclusions In this study, we provide an engineered lymphatic vessel platform that can generate four different flow regimes and reveal the roles of interstitial flow and VEGF-A/C for lymphatic sprouting and discontinuous junction formation. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00780-0.
Collapse
Affiliation(s)
- Isabelle S. Ilan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
- College of Human Ecology, Cornell University, Ithaca, NY 14853 USA
| | - Aria R. Yslas
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Yansong Peng
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Renhao Lu
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, College of Engineering, Cornell University, 302 Weill Hall, 237 Tower Road, Ithaca, NY 14853 USA
| |
Collapse
|
27
|
Buskin A, Scott E, Nelson R, Gaughan L, Robson CN, Heer R, Hepburn AC. Engineering prostate cancer in vitro: what does it take? Oncogene 2023; 42:2417-2427. [PMID: 37438470 PMCID: PMC10403358 DOI: 10.1038/s41388-023-02776-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023]
Abstract
A key challenge in the clinical management and cause of treatment failure of prostate cancer (PCa) is its molecular, cellular and clinical heterogeneity. Modelling systems that fully recapitulate clinical diversity and resistant phenotypes are urgently required for the development of successful personalised PCa therapies. The advent of the three-dimensional (3D) organoid model has revolutionised preclinical cancer research through reflecting heterogeneity and offering genomic and environmental manipulation that has opened up unparalleled opportunities for applications in disease modelling, high-throughput drug screening and precision medicine. Despite these remarkable achievements of organoid technology, several shortcomings in emulating the complex tumor microenvironment and dynamic process of metastasis as well as the epigenome profile limit organoids achieving true in vivo functionality. Technological advances in tissue engineering have enabled the development of innovative tools to facilitate the design of improved 3D cancer models. In this review, we highlight the current in vitro 3D PCa models with a special focus on organoids and discuss engineering approaches to create more physiologically relevant PCa organoid models and maximise their translational relevance that ultimately will help to realise the transformational power of precision medicine.
Collapse
Affiliation(s)
- Adriana Buskin
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Emma Scott
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Ryan Nelson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Luke Gaughan
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Craig N Robson
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rakesh Heer
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| | - Anastasia C Hepburn
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Paul O'Gorman Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
28
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
29
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
30
|
Visalakshan RM, Lowrey MK, Sousa MGC, Helms HR, Samiea A, Schutt CE, Moreau JM, Bertassoni LE. Opportunities and challenges to engineer 3D models of tumor-adaptive immune interactions. Front Immunol 2023; 14:1162905. [PMID: 37081897 PMCID: PMC10110941 DOI: 10.3389/fimmu.2023.1162905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Augmenting adaptive immunity is a critical goal for developing next-generation cancer therapies. T and B cells infiltrating the tumor dramatically influence cancer progression through complex interactions with the local microenvironment. Cancer cells evade and limit these immune responses by hijacking normal immunologic pathways. Current experimental models using conventional primary cells, cell lines, or animals have limitations for studying cancer-immune interactions directly relevant to human biology and clinical translation. Therefore, engineering methods to emulate such interplay at local and systemic levels are crucial to expedite the development of better therapies and diagnostic tools. In this review, we discuss the challenges, recent advances, and future directions toward engineering the tumor-immune microenvironment (TME), including key elements of adaptive immunity. We first offer an overview of the recent research that has advanced our understanding of the role of the adaptive immune system in the tumor microenvironment. Next, we discuss recent developments in 3D in-vitro models and engineering approaches that have been used to study the interaction of cancer and stromal cells with B and T lymphocytes. We summarize recent advancement in 3D bioengineering and discuss the need for 3D tumor models that better incorporate elements of the complex interplay of adaptive immunity and the tumor microenvironment. Finally, we provide a perspective on current challenges and future directions for modeling cancer-immune interactions aimed at identifying new biological targets for diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rahul M. Visalakshan
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Mary K. Lowrey
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Mauricio G. C. Sousa
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Haylie R. Helms
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Abrar Samiea
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Josh M. Moreau
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Department of Dermatology, Oregon Health and Science University, Portland, OR, United States
| | - Luiz E. Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, United States
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
31
|
Virgilio T, Bordini J, Cascione L, Sartori G, Latino I, Molina Romero D, Leoni C, Akhmedov M, Rinaldi A, Arribas AJ, Morone D, Seyed Jafari SM, Bersudsky M, Ottolenghi A, Kwee I, Chiaravalli AM, Sessa F, Hunger RE, Bruno A, Mortara L, Voronov E, Monticelli S, Apte RN, Bertoni F, Gonzalez SF. Subcapsular Sinus Macrophages Promote Melanoma Metastasis to the Sentinel Lymph Nodes via an IL1α-STAT3 Axis. Cancer Immunol Res 2022; 10:1525-1541. [PMID: 36206577 PMCID: PMC9716256 DOI: 10.1158/2326-6066.cir-22-0225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/18/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
During melanoma metastasis, tumor cells originating in the skin migrate via lymphatic vessels to the sentinel lymph node (sLN). This process facilitates tumor cell spread across the body. Here, we characterized the innate inflammatory response to melanoma in the metastatic microenvironment of the sLN. We found that macrophages located in the subcapsular sinus (SS) produced protumoral IL1α after recognition of tumoral antigens. Moreover, we confirmed that the elimination of LN macrophages or the administration of an IL1α-specific blocking antibody reduced metastatic spread. To understand the mechanism of action of IL1α in the context of the sLN microenvironment, we applied single-cell RNA sequencing to microdissected metastases obtained from animals treated with the IL1α-specific blocking antibody. Among the different pathways affected, we identified STAT3 as one of the main targets of IL1α signaling in metastatic tumor cells. Moreover, we found that the antitumoral effect of the anti-IL1α was not mediated by lymphocytes because Il1r1 knockout mice did not show significant differences in metastasis growth. Finally, we found a synergistic antimetastatic effect of the combination of IL1α blockade and STAT3 inhibition with stattic, highlighting a new immunotherapy approach to preventing melanoma metastasis.
Collapse
Affiliation(s)
- Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Joy Bordini
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,GenomSys SA, Lugano, Switzerland
| | - Luciano Cascione
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Giulio Sartori
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Daniel Molina Romero
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Graduate School Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristina Leoni
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Murodzhon Akhmedov
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alberto J. Arribas
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - S. Morteza Seyed Jafari
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aner Ottolenghi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ivo Kwee
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,BigOmics Analytics, Lugano, Switzerland
| | - Anna Maria Chiaravalli
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, ASST dei Sette Laghi, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Robert E. Hunger
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy.,Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Silvia Monticelli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Francesco Bertoni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.,Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Santiago F. Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Corresponding Author: Santiago F. Gonzalez, Institute for Research in Biomedicine, via Francesco Chiesa 5. CH-6500 Bellinzona. Switzerland. Phone: +41 58 666 7226; E-mail:
| |
Collapse
|
32
|
Bordy S, Byun J, Poulikakos LV. Nanophotonic materials: enabling targeted cancer diagnostics and therapeutics with light. Curr Opin Chem Eng 2022. [DOI: 10.1016/j.coche.2022.100852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
33
|
Zhang T, Jia Y, Yu Y, Zhang B, Xu F, Guo H. Targeting the tumor biophysical microenvironment to reduce resistance to immunotherapy. Adv Drug Deliv Rev 2022; 186:114319. [PMID: 35545136 DOI: 10.1016/j.addr.2022.114319] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 02/06/2023]
Abstract
Immunotherapy based on immune checkpoint inhibitors has evolved into a new pillar of cancer treatment in clinics, but dealing with treatment resistance (either primary or acquired) is a major challenge. The tumor microenvironment (TME) has a substantial impact on the pathological behaviors and treatment response of many cancers. The biophysical clues in TME have recently been considered as important characteristics of cancer. Furthermore, there is mounting evidence that biophysical cues in TME play important roles in each step of the cascade of cancer immunotherapy that synergistically contribute to immunotherapy resistance. In this review, we summarize five main biophysical cues in TME that affect resistance to immunotherapy: extracellular matrix (ECM) structure, ECM stiffness, tumor interstitial fluid pressure (IFP), solid stress, and vascular shear stress. First, the biophysical factors involved in anti-tumor immunity and therapeutic antibody delivery processes are reviewed. Then, the causes of these five biophysical cues and how they contribute to immunotherapy resistance are discussed. Finally, the latest treatment strategies that aim to improve immunotherapy efficacy by targeting these biophysical cues are shared. This review highlights the biophysical cues that lead to immunotherapy resistance, also supplements their importance in related technologies for studying TME biophysical cues in vitro and therapeutic strategies targeting biophysical cues to improve the effects of immunotherapy.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuanbo Jia
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yang Yu
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an 710061, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
34
|
Hammel JH, Zatorski JM, Cook SR, Pompano RR, Munson JM. Engineering in vitro immune-competent tissue models for testing and evaluation of therapeutics. Adv Drug Deliv Rev 2022; 182:114111. [PMID: 35031388 PMCID: PMC8908413 DOI: 10.1016/j.addr.2022.114111] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Advances in 3D cell culture, microscale fluidic control, and cellular analysis have enabled the development of more physiologically-relevant engineered models of human organs with precise control of the cellular microenvironment. Engineered models have been used successfully to answer fundamental biological questions and to screen therapeutics, but these often neglect key elements of the immune system. There are immune elements in every tissue that contribute to healthy and diseased states. Including immune function will be essential for effective preclinical testing of therapeutics for inflammatory and immune-modulated diseases. In this review, we first discuss the key components to consider in designing engineered immune-competent models in terms of physical, chemical, and biological cues. Next, we review recent applications of models of immunity for screening therapeutics for cancer, preclinical evaluation of engineered T cells, modeling autoimmunity, and screening vaccine efficacy. Future work is needed to further recapitulate immune responses in engineered models for the most informative therapeutic screening and evaluation.
Collapse
Affiliation(s)
- Jennifer H. Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Sophie R. Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA,Department of Biomedical Engineering, University of Virginia; Charlottesville, Virginia 22904, USA,Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| |
Collapse
|
35
|
Shou Y, Johnson SC, Quek YJ, Li X, Tay A. Integrative lymph node-mimicking models created with biomaterials and computational tools to study the immune system. Mater Today Bio 2022; 14:100269. [PMID: 35514433 PMCID: PMC9062348 DOI: 10.1016/j.mtbio.2022.100269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
The lymph node (LN) is a vital organ of the lymphatic and immune system that enables timely detection, response, and clearance of harmful substances from the body. Each LN comprises of distinct substructures, which host a plethora of immune cell types working in tandem to coordinate complex innate and adaptive immune responses. An improved understanding of LN biology could facilitate treatment in LN-associated pathologies and immunotherapeutic interventions, yet at present, animal models, which often have poor physiological relevance, are the most popular experimental platforms. Emerging biomaterial engineering offers powerful alternatives, with the potential to circumvent limitations of animal models, for in-depth characterization and engineering of the lymphatic and adaptive immune system. In addition, mathematical and computational approaches, particularly in the current age of big data research, are reliable tools to verify and complement biomaterial works. In this review, we first discuss the importance of lymph node in immunity protection followed by recent advances using biomaterials to create in vitro/vivo LN-mimicking models to recreate the lymphoid tissue microstructure and microenvironment, as well as to describe the related immuno-functionality for biological investigation. We also explore the great potential of mathematical and computational models to serve as in silico supports. Furthermore, we suggest how both in vitro/vivo and in silico approaches can be integrated to strengthen basic patho-biological research, translational drug screening and clinical personalized therapies. We hope that this review will promote synergistic collaborations to accelerate progress of LN-mimicking systems to enhance understanding of immuno-complexity.
Collapse
Key Words
- ABM, agent-based model
- APC, antigen-presenting cell
- BV, blood vessel
- Biomaterials
- CPM, Cellular Potts model
- Computational models
- DC, dendritic cell
- ECM, extracellular matrix
- FDC, follicular dendritic cell
- FRC, fibroblastic reticular cell
- Immunotherapy
- LEC, lymphatic endothelial cell
- LN, lymph node
- LV, lymphatic vessel
- Lymph node
- Lymphatic system
- ODE, ordinary differential equation
- PDE, partial differential equation
- PDMS, polydimethylsiloxane
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Sarah C. Johnson
- Department of Bioengineering, Stanford University, CA, 94305, USA
- Department of Bioengineering, Imperial College London, South Kensington, SW72AZ, UK
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, 138648, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
36
|
Giantesio G, Girelli A, Musesti A. A Mathematical Description of the Flow in a Spherical Lymph Node. Bull Math Biol 2022; 84:142. [PMID: 36318334 PMCID: PMC9626437 DOI: 10.1007/s11538-022-01103-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 11/13/2022]
Abstract
The motion of the lymph has a very important role in the immune system, and it is influenced by the porosity of the lymph nodes: more than 90% takes the peripheral path without entering the lymphoid compartment. In this paper, we construct a mathematical model of a lymph node assumed to have a spherical geometry, where the subcapsular sinus is a thin spherical shell near the external wall of the lymph node and the core is a porous material describing the lymphoid compartment. For the mathematical formulation, we assume incompressibility and we use Stokes together with Darcy-Brinkman equation for the flow of the lymph. Thanks to the hypothesis of axisymmetric flow with respect to the azimuthal angle and the use of the stream function approach, we find an explicit solution for the fully developed pulsatile flow in terms of Gegenbauer polynomials. A selected set of plots is provided to show the trend of motion in the case of physiological parameters. Then, a finite element simulation is performed and it is compared with the explicit solution.
Collapse
Affiliation(s)
- Giulia Giantesio
- grid.8142.f0000 0001 0941 3192Dipartimento di Matematica e Fisica “N. Tartaglia”, Università Cattolica del Sacro Cuore, Brescia, Italy
| | - Alberto Girelli
- grid.7563.70000 0001 2174 1754Dipartimento di Matematica e Applicazioni, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Alessandro Musesti
- grid.8142.f0000 0001 0941 3192Dipartimento di Matematica e Fisica “N. Tartaglia”, Università Cattolica del Sacro Cuore, Brescia, Italy
| |
Collapse
|
37
|
Alderfer L, Hall E, Hanjaya-Putra D. Harnessing biomaterials for lymphatic system modulation. Acta Biomater 2021; 133:34-45. [PMID: 34118451 PMCID: PMC9113193 DOI: 10.1016/j.actbio.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. To bridge this knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease processes, including cancer metastasis and inflammation. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. This review article focuses on recent advances in (i) biomaterials used for lymphatic vessel formation, (ii) models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) and strategies for drug delivery via the lymphatic system. Finally, several challenges regarding adopting biomaterials for immunomodulation and future perspectives are discussed. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. This review article focuses on recent progresses in biomaterials to investigate the lymphatic system and to provide novel understanding into complex disease states. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.
Collapse
|
38
|
Shanti A, Hallfors N, Petroianu GA, Planelles L, Stefanini C. Lymph Nodes-On-Chip: Promising Immune Platforms for Pharmacological and Toxicological Applications. Front Pharmacol 2021; 12:711307. [PMID: 34483920 PMCID: PMC8415712 DOI: 10.3389/fphar.2021.711307] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Organs-on-chip are gaining increasing attention as promising platforms for drug screening and testing applications. However, lymph nodes-on-chip options remain limited although the lymph node is one of the main determinants of the immunotoxicity of newly developed pharmacological drugs. In this review, we describe existing biomimetic lymph nodes-on-chip, their design, and their physiological relevance to pharmacology and shed the light on future directions associated with lymph node-on-chip design and implementation in drug discovery and development.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Georg A Petroianu
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Lourdes Planelles
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|