1
|
Gaebler D, Hachey SJ, Hughes CCW. Microphysiological systems as models for immunologically 'cold' tumors. Front Cell Dev Biol 2024; 12:1389012. [PMID: 38711620 PMCID: PMC11070549 DOI: 10.3389/fcell.2024.1389012] [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: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024] Open
Abstract
The tumor microenvironment (TME) is a diverse milieu of cells including cancerous and non-cancerous cells such as fibroblasts, pericytes, endothelial cells and immune cells. The intricate cellular interactions within the TME hold a central role in shaping the dynamics of cancer progression, influencing pivotal aspects such as tumor initiation, growth, invasion, response to therapeutic interventions, and the emergence of drug resistance. In immunologically 'cold' tumors, the TME is marked by a scarcity of infiltrating immune cells, limited antigen presentation in the absence of potent immune-stimulating signals, and an abundance of immunosuppressive factors. While strategies targeting the TME as a therapeutic avenue in 'cold' tumors have emerged, there is a pressing need for novel approaches that faithfully replicate the complex cellular and non-cellular interactions in order to develop targeted therapies that can effectively stimulate immune responses and improve therapeutic outcomes in patients. Microfluidic devices offer distinct advantages over traditional in vitro 3D co-culture models and in vivo animal models, as they better recapitulate key characteristics of the TME and allow for precise, controlled insights into the dynamic interplay between various immune, stromal and cancerous cell types at any timepoint. This review aims to underscore the pivotal role of microfluidic systems in advancing our understanding of the TME and presents current microfluidic model systems that aim to dissect tumor-stromal, tumor-immune and immune-stromal cellular interactions in various 'cold' tumors. Understanding the intricacies of the TME in 'cold' tumors is crucial for devising effective targeted therapies to reinvigorate immune responses and overcome the challenges of current immunotherapy approaches.
Collapse
Affiliation(s)
- Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Stephanie J. Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
2
|
Hachey SJ, Hatch CJ, Gaebler D, Mocherla A, Nee K, Kessenbrock K, Hughes CCW. Targeting tumor-stromal interactions in triple-negative breast cancer using a human vascularized micro-tumor model. Breast Cancer Res 2024; 26:5. [PMID: 38183074 PMCID: PMC10768273 DOI: 10.1186/s13058-023-01760-y] [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: 07/30/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive with limited available treatments. Stromal cells in the tumor microenvironment (TME) are crucial in TNBC progression; however, understanding the molecular basis of stromal cell activation and tumor-stromal crosstalk in TNBC is limited. To investigate therapeutic targets in the TNBC stromal niche, we used an advanced human in vitro microphysiological system called the vascularized micro-tumor (VMT). Using single-cell RNA sequencing, we revealed that normal breast tissue stromal cells activate neoplastic signaling pathways in the TNBC TME. By comparing interactions in VMTs with clinical data, we identified therapeutic targets at the tumor-stromal interface with potential clinical significance. Combining treatments targeting Tie2 signaling with paclitaxel resulted in vessel normalization and increased efficacy of paclitaxel in the TNBC VMT. Dual inhibition of HER3 and Akt also showed efficacy against TNBC. These data demonstrate the potential of inducing a favorable TME as a targeted therapeutic approach in TNBC.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
| | | | - Daniela Gaebler
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Aneela Mocherla
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Kevin Nee
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Christopher C W Hughes
- Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
3
|
Campo H, Zha D, Pattarawat P, Colina J, Zhang D, Murphy A, Yoon J, Russo A, Rogers HB, Lee HC, Zhang J, Trotter K, Wagner S, Ingram A, Pavone ME, Dunne SF, Boots CE, Urbanek M, Xiao S, Burdette JE, Woodruff TK, Kim JJ. A new tissue-agnostic microfluidic device to model physiology and disease: the lattice platform. LAB ON A CHIP 2023; 23:4821-4833. [PMID: 37846545 PMCID: PMC11181516 DOI: 10.1039/d3lc00378g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
To accurately phenocopy human biology in vitro, researchers have been reducing their dependence on standard, static two-dimensional (2D) cultures and instead are moving towards three-dimensional (3D) and/or multicellular culture techniques. While these culture innovations are becoming more commonplace, there is a growing body of research that illustrates the benefits and even necessity of recapitulating the dynamic flow of nutrients, gas, waste exchange and tissue interactions that occur in vivo. However, cost and engineering complexity are two main factors that hinder the adoption of these technologies and incorporation into standard laboratory workflows. We developed LATTICE, a plug-and-play microfluidic platform able to house up to eight large tissue or organ models that can be cultured individually or in an interconnected fashion. The functionality of the platform to model both healthy and diseased tissue states was demonstrated using 3D cultures of reproductive tissues including murine ovarian tissues and human fallopian tube explants (hFTE). When exogenously exposed to pathological doses of gonadotropins and androgens to mimic the endocrinology of polycystic ovarian syndrome (PCOS), subsequent ovarian follicle development, hormone production and ovulation copied key features of this endocrinopathy. Further, hFTE cilia beating decreased significantly only when experiencing continuous media exchanges. We were then able to endogenously recreate this phenotype on the platform by dynamically co-culturing the PCOS ovary and hFTE. LATTICE was designed to be customizable with flexibility in 3D culture formats and can serve as a powerful automated tool to enable the study of tissue and cellular dynamics in health and disease in all fields of research.
Collapse
Affiliation(s)
- Hannes Campo
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Didi Zha
- Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Pawat Pattarawat
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Jose Colina
- Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Delong Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Alina Murphy
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Julia Yoon
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Angela Russo
- Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hunter B Rogers
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Hoi Chang Lee
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Jiyang Zhang
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Katy Trotter
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Sarah Wagner
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Asia Ingram
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Sara Fernandez Dunne
- High-throughput Analysis Laboratory, Northwestern University, Evanston, IL 60628, USA
| | - Christina E Boots
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, Center for Biomolecular Science, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- Department of Obstetrics and Gynecology, Michigan State University, East Lansing, MI 48824, USA
| | - J Julie Kim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
4
|
Hachey SJ, Sobrino A, Lee JG, Jafari MD, Klempner SJ, Puttock EJ, Edwards RA, Lowengrub JS, Waterman ML, Zell JA, Hughes CCW. A human vascularized microtumor model of patient-derived colorectal cancer recapitulates clinical disease. Transl Res 2023; 255:97-108. [PMID: 36481562 PMCID: PMC10593408 DOI: 10.1016/j.trsl.2022.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Accurately modeling tumor biology and testing novel therapies on patient-derived cells is critically important to developing therapeutic regimens personalized to a patient's specific disease. The vascularized microtumor (VMT), or "tumor-on-a-chip," is a physiologic preclinical cancer model that incorporates key features of the native human tumor microenvironment within a transparent microfluidic platform, allowing rapid drug screening in vitro. Herein we optimize methods for generating patient-derived VMT (pVMT) using fresh colorectal cancer (CRC) biopsies and surgical resections to test drug sensitivities at the individual patient level. In response to standard chemotherapy and TGF-βR1 inhibition, we observe heterogeneous responses between pVMT derived from 6 patient biopsies, with the pVMT recapitulating tumor growth, histological features, metabolic heterogeneity, and drug responses of actual CRC tumors. Our results suggest that a translational infrastructure providing rapid information from patient-derived tumor cells in the pVMT, as established in this study, will support efforts to improve patient outcomes.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Irvine Department of Molecular Biology and Biochemistry, University of California, Irvine, California
| | - Agua Sobrino
- Irvine Department of Molecular Biology and Biochemistry, University of California, Irvine, California
| | - John G Lee
- Irvine School of Medicine, University of California, Irvine, California
| | | | | | - Eric J Puttock
- Irvine Department of Mathematics, University of California, Irvine, California
| | - Robert A Edwards
- Irvine School of Medicine, University of California, Irvine, California
| | - John S Lowengrub
- Irvine Department of Mathematics, University of California, Irvine, California
| | - Marian L Waterman
- Irvine Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| | - Jason A Zell
- Irvine School of Medicine, University of California, Irvine, California
| | - Christopher C W Hughes
- Irvine Department of Molecular Biology and Biochemistry, University of California, Irvine, California; Irvine Department of Biomedical Engineering, University of California, Irvine, California.
| |
Collapse
|
5
|
Caballero D, Reis RL, Kundu SC. Engineering Patient-on-a-Chip Models for Personalized Cancer Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1230:43-64. [PMID: 32285364 DOI: 10.1007/978-3-030-36588-2_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traditional in vitro and in vivo models typically used in cancer research have demonstrated a low predictive power for human response. This leads to high attrition rates of new drugs in clinical trials, which threaten cancer patient prognosis. Tremendous efforts have been directed towards the development of a new generation of highly predictable pre-clinical models capable to reproduce in vitro the biological complexity of the human body. Recent advances in nanotechnology and tissue engineering have enabled the development of predictive organs-on-a-chip models of cancer with advanced capabilities. These models can reproduce in vitro the complex three-dimensional physiology and interactions that occur between organs and tissues in vivo, offering multiple advantages when compared to traditional models. Importantly, these models can be tailored to the biological complexity of individual cancer patients resulting into biomimetic and personalized cancer patient-on-a-chip platforms. The individualized models provide a more accurate and physiological environment to predict tumor progression on patients and their response to drugs. In this chapter, we describe the latest advances in the field of cancer patient-on-a-chip, and discuss about their main applications and current challenges. Overall, we anticipate that this new paradigm in cancer in vitro models may open up new avenues in the field of personalized - cancer - medicine, which may allow pharmaceutical companies to develop more efficient drugs, and clinicians to apply patient-specific therapies.
Collapse
Affiliation(s)
- David Caballero
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal. .,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal.,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, Guimarães, Portugal.,ICVS 3Bs PT Government Associate Lab, Braga, Guimarães, Portugal
| |
Collapse
|
6
|
Motallebnejad P, Thomas A, Swisher SL, Azarin SM. An isogenic hiPSC-derived BBB-on-a-chip. BIOMICROFLUIDICS 2019; 13:064119. [PMID: 31768205 PMCID: PMC6874510 DOI: 10.1063/1.5123476] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/07/2019] [Indexed: 05/03/2023]
Abstract
The blood-brain barrier (BBB) is composed of brain microvascular endothelial cells (BMECs) that regulate brain homeostasis, and astrocytes within the brain are involved in the maintenance of the BBB or modulation of its integrity in disease states via secreted factors. A major challenge in modeling the normal or diseased BBB is that conventional in vitro models lack either the physiological complexity of the BBB or key functional features such as formation of a sufficiently tight barrier. In this study, we utilized human induced pluripotent stem cell (hiPSC)-derived BMECs in a BBB-on-a-chip device that supports flow and coculture with an astrocyte-laden 3D hydrogel. The BMECs are separated from the hydrogel by a porous membrane with either 0.4 or 8.0 μm pore size, making the device suitable for studying the transport of molecules or cells, respectively, across the BBB. In addition, all cells seeded in the device are differentiated from the same hiPSC line, which could enable genetic and rare disease modeling. Formation of a confluent BMEC barrier was confirmed by immunocytochemistry of tight junction proteins and measurement of fluorescein permeability. Integrity of the barrier was further assessed by performing impedance spectroscopy in the device. Finally, the ability of this device to recapitulate a disease model of BBB disruption was demonstrated, with apical addition of TGF-β1 leading to transendothelial electrical resistance reduction and indicators of astrocyte activation. These results demonstrate the utility of the fabricated device for a broad range of applications such as drug screening and mechanistic studies of BBB disruption.
Collapse
Affiliation(s)
- Pedram Motallebnejad
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Andrew Thomas
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Sarah L. Swisher
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Samira M. Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
7
|
Ballesteros Hernando J, Ramos Gómez M, Díaz Lantada A. Modeling Living Cells Within Microfluidic Systems Using Cellular Automata Models. Sci Rep 2019; 9:14886. [PMID: 31624307 PMCID: PMC6797720 DOI: 10.1038/s41598-019-51494-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/02/2019] [Indexed: 11/25/2022] Open
Abstract
Several computational models, both continuum and discrete, allow for the simulation of collective cell behaviors in connection with challenges linked to disease modeling and understanding. Normally, discrete cell modelling employs quasi-infinite or boundary-less 2D lattices, hence modeling collective cell behaviors in Petri dish-like environments. The advent of lab- and organ-on-a-chip devices proves that the information obtained from 2D cell cultures, upon Petri dishes, differs importantly from the results obtained in more biomimetic micro-fluidic environments, made of interconnected chambers and channels. However, discrete cell modelling within lab- and organ-on-a-chip devices, to our knowledge, is not yet found in the literature, although it may prove useful for designing and optimizing these types of systems. Consequently, in this study we focus on the establishment of a direct connection between the computer-aided designs (CAD) of microfluidic systems, especially labs- and organs-on-chips (and their multi-chamber and multi-channel structures), and the lattices for discrete cell modeling approaches aimed at the simulation of collective cell interactions, whose boundaries are defined directly from the CAD models. We illustrate the proposal using a quite straightforward cellular automata model, apply it to simulating cells with different growth rates, within a selected set of microsystem designs, and validate it by tuning the growth rates with the support of cell culture experiments and by checking the results with a real microfluidic system.
Collapse
Affiliation(s)
- Julia Ballesteros Hernando
- Laboratorio de Desarrollo de Productos, Departamento de Ingeniería Mecánica, Universidad Politécnica de Madrid, c/José Gutiérrez Abascal 2, 28006, Madrid, Spain.,Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Parque Científico y Tecnológico, M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Milagros Ramos Gómez
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Parque Científico y Tecnológico, M40, Km. 38, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Andrés Díaz Lantada
- Laboratorio de Desarrollo de Productos, Departamento de Ingeniería Mecánica, Universidad Politécnica de Madrid, c/José Gutiérrez Abascal 2, 28006, Madrid, Spain.
| |
Collapse
|
8
|
Renggli K, Rousset N, Lohasz C, Nguyen OTP, Hierlemann A. Integrated Microphysiological Systems: Transferable Organ Models and Recirculating Flow. ADVANCED BIOSYSTEMS 2019; 3:e1900018. [PMID: 32627410 PMCID: PMC7610576 DOI: 10.1002/adbi.201900018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.
Collapse
Affiliation(s)
- Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | | | |
Collapse
|
9
|
Polini A, Del Mercato LL, Barra A, Zhang YS, Calabi F, Gigli G. Towards the development of human immune-system-on-a-chip platforms. Drug Discov Today 2019; 24:517-525. [PMID: 30312743 PMCID: PMC6440212 DOI: 10.1016/j.drudis.2018.10.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 01/22/2023]
Abstract
Organ-on-a-chip (OoCs) platforms could revolutionize drug discovery and might ultimately become essential tools for precision therapy. Although many single-organ and interconnected systems have been described, the immune system has been comparatively neglected, despite its pervasive role in the body and the trend towards newer therapeutic products (i.e., complex biologics, nanoparticles, immune checkpoint inhibitors, and engineered T cells) that often cause, or are based on, immune reactions. In this review, we recapitulate some distinctive features of the immune system before reviewing microfluidic devices that mimic lymphoid organs or other organs and/or tissues with an integrated immune system component.
Collapse
Affiliation(s)
- Alessandro Polini
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy.
| | - Loretta L Del Mercato
- CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Adriano Barra
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; INFN, Sezione di Lecce, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; INdAM (GNFM), Sezione di Lecce, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Franco Calabi
- CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| | - Giuseppe Gigli
- Dipartimento di Matematica e Fisica E. De Giorgi, University of Salento, Campus Ecotekne, via Monteroni, 73100, Lecce, Italy; CNR NANOTEC - Institute of Nanotechnology c/o Campus Ecotekne, via Monteroni, 73100, Lecce, Italy
| |
Collapse
|
10
|
Rudmann DG. The Emergence of Microphysiological Systems (Organs-on-chips) as Paradigm-changing Tools for Toxicologic Pathology. Toxicol Pathol 2018; 47:4-10. [PMID: 30407146 DOI: 10.1177/0192623318809065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Microphysiological systems (MPS), commonly known as organs-on-chips, are a rapidly advancing technology that promises to impact many areas of medical and toxicological pathology. In this minireview, the history of MPS and its potential utility in safety assessment are described with the toxicologic pathologist in mind. Several MPS development focus areas are defined, and recent progress in the area is highlighted. MPS will likely become an important tool for the toxicologic pathologist as part of our role in the safety assessment process within the pharmaceutical, biotechnology, medical device, and cosmetic and agrichemical industries.
Collapse
|
11
|
Abstract
Over the past six decades the inflation-adjusted cost to bring a new drug to market has been increasing constantly and doubles every 9 years - now reaching in excess of $2.5 billion. Overall, the likelihood of FDA approval for a drug (any disease indication) that has entered phase I clinical trials is a mere 9.6%, with the approval rate for oncology far below average at only 5.1%. Lack of efficacy or toxicity is often not revealed until the later stages of clinical trials, despite promising preclinical data. This indicates that the current in vitro systems for drug screening need to be improved for better predictability of in vivo outcomes. Microphysiological systems (MPS), or bioengineered 3D microfluidic tissue and organ constructs that mimic physiological and pathological processes in vitro, can be leveraged across preclinical research and clinical trial stages to transform drug development and clinical management for a range of diseases. Here we review the current state-of-the-art in 3D tissue-engineering models developed for cancer research, with a focus on tumor-on-a-chip, or tumor chip, models. From our viewpoint, tumor chip systems can advance innovative medicine to ameliorate the high failure rates in anti-cancer drug development and clinical treatment.
Collapse
Affiliation(s)
- Stephanie J Hachey
- Department of Molecular Biology & Biochemistry, University of California, Irvine, CA 92697, USA.
| | | |
Collapse
|
12
|
Li X, George SM, Vernetti L, Gough AH, Taylor DL. A glass-based, continuously zonated and vascularized human liver acinus microphysiological system (vLAMPS) designed for experimental modeling of diseases and ADME/TOX. LAB ON A CHIP 2018; 18:2614-2631. [PMID: 30063238 PMCID: PMC6113686 DOI: 10.1039/c8lc00418h] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The vLAMPS is a human, biomimetic liver MPS, in which the ECM and cell seeding of the intermediate layer prior to assembly, simplifies construction of the model and makes the platform user-friendly. This primarily glass microfluidic device is optimal for real-time imaging, while minimizing the binding of hydrophobic drugs/biologics to the materials that constitute the device. The assembly of the three layered device with primary human hepatocytes and liver sinusoidal endothelial cells (LSECs), and human cell lines for stellate and Kupffer cells, creates a vascular channel separated from the hepatic channel (chamber) by a porous membrane that allows communication between channels, recapitulating the 3D structure of the liver acinus. The vascular channel can be used to deliver drugs, immune cells, as well as various circulating cells and other factors to a stand-alone liver MPS and/or to couple the liver MPS to other organ MPS. We have successfully created continuous oxygen zonation by controlling the flow rates of media in the distinct vascular and hepatic channels and validated the computational modeling of zonation with oxygen sensitive and insensitive beads. This allows the direct investigation of the role of zonation in physiology, toxicology and disease progression. The vascular channel is lined with human LSECs, recapitulating partial immunologic functions within the liver sinusoid, including the activation of LSECs, promoting the binding of polymorphonuclear leukocytes (PMNs) followed by transmigration into the hepatic chamber. The vLAMPS is a valuable platform to investigate the functions of the healthy and diseased human liver using all primary human cell types and/or iPSC-derived cells.
Collapse
Affiliation(s)
- Xiang Li
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | | | | | | | | |
Collapse
|