1
|
Ghoshal D, Petersen I, Ringquist R, Kramer L, Bhatia E, Hu T, Richard A, Park R, Corbin J, Agarwal S, Thomas A, Ramirez S, Tharayil J, Downey E, Ketchum F, Ochal A, Sonthi N, Lonial S, Kochenderfer JN, Tran R, Zhu M, Lam WA, Coskun AF, Roy K. Multi-niche human bone marrow on-a-chip for studying the interactions of adoptive CAR-T cell therapies with multiple myeloma. Biomaterials 2025; 316:123016. [PMID: 39709851 DOI: 10.1016/j.biomaterials.2024.123016] [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: 07/25/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Multiple myeloma (MM), a cancer of bone marrow plasma cells, is the second-most common hematological malignancy. However, despite immunotherapies like chimeric antigen receptor (CAR)-T cells, relapse is nearly universal. The bone marrow (BM) microenvironment influences how MM cells survive, proliferate, and resist treatment. Yet, it is unclear which BM niches give rise to MM pathophysiology. Here, we present a 3D microvascularized culture system, which models the endosteal and perivascular bone marrow niches, allowing us to study MM-stroma interactions in the BM niche and model responses to therapeutic CAR-T cells. We demonstrated the prolonged survival of cell line-based and patient-derived multiple myeloma cells within our in vitro system and successfully perfused in donor-matched CAR-T cells. We then measured T cell survival, differentiation, and cytotoxicity against MM cells using a variety of analysis techniques. Our MM-on-a-chip system could elucidate the role of the BM microenvironment in MM survival and therapeutic evasion and inform the rational design of next-generation therapeutics.
Collapse
Affiliation(s)
- Delta Ghoshal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ingrid Petersen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Rachel Ringquist
- The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Liana Kramer
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Eshant Bhatia
- The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; The George W. Woodruff Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Thomas Hu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ariane Richard
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Reda Park
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Jenna Corbin
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Savi Agarwal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Abel Thomas
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Department of Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Sebastian Ramirez
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Jacob Tharayil
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Emma Downey
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Frank Ketchum
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Abigail Ochal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Neha Sonthi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Reginald Tran
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mandy Zhu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ahmet F Coskun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; The Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Krishnendu Roy
- School of Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Kozalak G, Koşar A. Bone-on-a-Chip Systems for Hematological Cancers. BIOSENSORS 2025; 15:176. [PMID: 40136973 PMCID: PMC11940066 DOI: 10.3390/bios15030176] [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: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Hematological malignancies originating from blood, bone marrow, and lymph nodes include leukemia, lymphoma, and myeloma, which necessitate the use of a distinct chemotherapeutic approach. Drug resistance frequently complicates their treatment, highlighting the need for predictive tools to guide therapeutic decisions. Conventional 2D/3D cell cultures do not fully encompass in vivo criteria, and translating disease models from mice to humans proves challenging. Organ-on-a-chip technology presents an avenue to surmount genetic disparities between species, offering precise design, concurrent manipulation of various cell types, and extrapolation of data to human physiology. The development of bone-on-a-chip (BoC) systems is crucial for accurately representing the in vivo bone microenvironment, predicting drug responses for hematological cancers, mitigating drug resistance, and facilitating personalized therapeutic interventions. BoC systems for modeling hematological cancers and drug research can encompass intricate designs and integrated platforms for analyzing drug response data to simulate disease scenarios. This review provides a comprehensive examination of BoC systems applicable to modeling hematological cancers and visualizing drug responses within the intricate context of bone. It thoroughly discusses the materials pertinent to BoC systems, suitable in vitro techniques, the predictive capabilities of BoC systems in clinical settings, and their potential for commercialization.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
3
|
Bayona C, Olaizola-Rodrigo C, Sharko V, Ashrafi M, Barrio JD, Doblaré M, Monge R, Ochoa I, Oliván S. A Novel Multicompartment Barrier-Free Microfluidic Device Reveals the Impact of Extracellular Matrix Stiffening and Temozolomide on Immune-Tumor Interactions in Glioblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409229. [PMID: 39901479 DOI: 10.1002/smll.202409229] [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: 10/08/2024] [Revised: 01/17/2025] [Indexed: 02/05/2025]
Abstract
The immune system plays a crucial role in shaping the glioblastoma tumor microenvironment, characterized by its complexity and dynamic interactions. Understanding the tumor-immune crosstalk is essential for advancing cancer research and therapeutic development. Here, a novel multicompartment, barrier-free microfluidic device is presented that overcomes the limitations of existing models by enabling direct tumor-immune interactions without physical barriers, preserving natural immune cell infiltration. This platform supports the independent and simultaneous culture of tumor and immune cells, replicating the healthy-tumoral stroma interface, and allows investigating the effect of matrix stiffness and chemotherapy on both populations. The findings reveal that increased collagen concentration promotes tumor invasiveness while impairing immune cell infiltration. Additionally, temozolomide treatment reduces immune cell motility but enhances anti-tumor immune responses. These insights highlight the critical roles of extracellular matrix mechanics and chemotherapy in tumor progression and immune modulation, establishing this device as a powerful tool for studying glioblastoma-immune dynamics and evaluating therapeutic strategies.
Collapse
Affiliation(s)
- Clara Bayona
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- BEOnChip S.L., Zaragoza, 50018, Spain
| | - Vira Sharko
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Mehran Ashrafi
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| | - Jesús Del Barrio
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Manuel Doblaré
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Department of Organic Chemistry, Zaragoza, 50009, Spain
| | | | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-University of Zaragoza, Department of Organic Chemistry, Zaragoza, 50009, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, 50009, Spain
| |
Collapse
|
4
|
Ajikumar A, Lei KF. Microfluidic Technologies in Advancing Cancer Research. MICROMACHINES 2024; 15:1444. [PMID: 39770196 PMCID: PMC11677295 DOI: 10.3390/mi15121444] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
This review explores the significant role of microfluidic technologies in advancing cancer research, focusing on the below key areas: droplet-based microfluidics, organ-on-chip systems, paper-based microfluidics, electrokinetic chips, and microfluidic chips for the study of immune response. Droplet-based microfluidics allows precise manipulation of cells and three-dimensional microtissues, enabling high-throughput experiments that reveal insights into cancer cell migration, invasion, and drug resistance. Organ-on-chip systems replicate human organs to assess drug efficacy and toxicity, particularly in the liver, heart, kidney, gut, lung, and brain. Paper-based microfluidics offers an alternative approach to accomplish rapid diagnostics and cell- and tissue-based bioassays. Electrokinetic microfluidic chips offer precise control over cell positioning and behavior, facilitating drug screening and cellular studies. Immune response studies leverage real-time observation of interactions between immune and cancer cells, supporting the development of immunotherapies. These microfluidic advances are paving the way for personalized cancer treatments while addressing challenges of scalability, cost, and clinical integration.
Collapse
Affiliation(s)
- Arjun Ajikumar
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Kin Fong Lei
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Department of Electrical & Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
5
|
Offeddu GS, Cambria E, Shelton SE, Haase K, Wan Z, Possenti L, Nguyen HT, Gillrie MR, Hickman D, Knutson CG, Kamm RD. Personalized Vascularized Models of Breast Cancer Desmoplasia Reveal Biomechanical Determinants of Drug Delivery to the Tumor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402757. [PMID: 39041892 PMCID: PMC11481247 DOI: 10.1002/advs.202402757] [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: 03/15/2024] [Revised: 07/01/2024] [Indexed: 07/24/2024]
Abstract
Desmoplasia in breast cancer leads to heterogeneity in physical properties of the tissue, resulting in disparities in drug delivery and treatment efficacy among patients, thus contributing to high disease mortality. Personalized in vitro breast cancer models hold great promise for high-throughput testing of therapeutic strategies to normalize the aberrant microenvironment in a patient-specific manner. Here, tumoroids assembled from breast cancer cell lines (MCF7, SKBR3, and MDA-MB-468) and patient-derived breast tumor cells (TCs) cultured in microphysiological systems including perfusable microvasculature reproduce key aspects of stromal and vascular dysfunction causing impaired drug delivery. Models containing SKBR3 and MDA-MB-468 tumoroids show higher stromal hyaluronic acid (HA) deposition, vascular permeability, interstitial fluid pressure (IFP), and degradation of vascular HA relative to models containing MCF7 tumoroids or models without tumoroids. Interleukin 8 (IL8) secretion is found responsible for vascular dysfunction and loss of vascular HA. Interventions targeting IL8 or stromal HA normalize vascular permeability, perfusion, and IFP, and ultimately enhance drug delivery and TC death in response to perfusion with trastuzumab and cetuximab. Similar responses are observed in patient-derived models. These microphysiological systems can thus be personalized by using patient-derived cells and can be applied to discover new molecular therapies for the normalization of the tumor microenvironment.
Collapse
Affiliation(s)
- Giovanni S. Offeddu
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Elena Cambria
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah E. Shelton
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Kristina Haase
- European Molecular Biology LaboratoryEuropean Molecular Biology Laboratory BarcelonaBarcelona08003Spain
| | - Zhengpeng Wan
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Luca Possenti
- LaBSDepartment of ChemistryMaterials and Chemical EngineeringPolitecnico di MilanoMilan20133Italy
- Data Science UnitFondazione IRCCS Istituto Nazionale dei TumoriMilan20133Italy
| | - Huu Tuan Nguyen
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark R. Gillrie
- Department of MedicineSnyder Institute for Chronic DiseasesUniversity of CalgaryCalgaryAlbertaT2N 2T9Canada
| | - Dean Hickman
- Amgen ResearchAmgen Inc.360 Binney StreetCambridgeMA02142USA
| | | | - Roger D. Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
6
|
Bonifácio ED, Araújo CA, Guimarães MV, de Souza MP, Lima TP, de Avelar Freitas BA, González-Torres LA. Computational model of the cancer necrotic core formation in a tumor-on-a-chip device. J Theor Biol 2024; 592:111893. [PMID: 38944380 DOI: 10.1016/j.jtbi.2024.111893] [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: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
The mechanisms underlying the formation of necrotic regions within avascular tumors are complex and poorly understood. In this paper, we investigate the formation of a necrotic core in a 3D tumor cell culture within a microfluidic device, considering oxygen, nutrients, and the microenvironment acidification by means of a computational-mathematical model. Our objective is to simulate cell processes, including proliferation and death inside a microfluidic device, according to the microenvironmental conditions. We employed approximation utilizing finite element models taking into account glucose, oxygen, and hydrogen ions diffusion, consumption and production, as well as cell proliferation, migration and death, addressing how tumor cells evolve under different conditions. The resulting mathematical model was examined under different scenarios, being capable of reproducing cell death and proliferation under different cell concentrations, and the formation of a necrotic core, in good agreement with experimental data reported in the literature. This approach not only advances our fundamental understanding of necrotic core formation but also provides a robust computational platform to study personalized therapeutic strategies, offering an important tool in cancer research and treatment design.
Collapse
Affiliation(s)
- Elton Diêgo Bonifácio
- Institute of Science and Technology - UFVJM, Diamantina, Brazil; Brazilian Reference Center for Assistive Technological Innovations (CINTESP.Br) - UFU, Uberlandia, Brazil.
| | - Cleudmar Amaral Araújo
- Brazilian Reference Center for Assistive Technological Innovations (CINTESP.Br) - UFU, Uberlandia, Brazil
| | | | - Márcio Peres de Souza
- Brazilian Reference Center for Assistive Technological Innovations (CINTESP.Br) - UFU, Uberlandia, Brazil
| | | | | | | |
Collapse
|
7
|
Peng AY, Lee BE. Microphysiological Systems for Cancer Immunotherapy Research and Development. Adv Biol (Weinh) 2024; 8:e2300077. [PMID: 37409385 PMCID: PMC10770294 DOI: 10.1002/adbi.202300077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
Cancer immunotherapy focuses on the use of patients' adaptive immune systems to combat cancer. In the past decade, FDA has approved many immunotherapy products for cancer patients who suffer from primary tumors, tumor relapse, and metastases. However, these immunotherapies still show resistance in many patients and often lead to inconsistent responses in patients due to variations in tumor genetic mutations and tumor immune microenvironment. Microfluidics-based organ-on-a-chip technologies or microphysiological systems have opened new ways that can provide relatively fast screening for personalized immunotherapy and help researchers and clinicians understand tumor-immune interactions in a patient-specific manner. They also have the potential to overcome the limitations of traditional drug screening and testing, given the models provide a more realistic 3D microenvironment with better controllability, reproducibility, and physiological relevance. This review focuses on the cutting-edge microphysiological organ-on-a-chip devices developed in recent years for studying cancer immunity and testing cancer immunotherapeutic agents, as well as some of the largest challenges of translating this technology to clinical applications in immunotherapy and personalized medicine.
Collapse
Affiliation(s)
- A. Yansong Peng
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - B. Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
8
|
Wan Y, Ding J, Jia Z, Hong Y, Tian G, Zheng S, Pan P, Wang J, Liang H. Current trends and research topics regarding organoids: A bibliometric analysis of global research from 2000 to 2023. Heliyon 2024; 10:e32965. [PMID: 39022082 PMCID: PMC11253259 DOI: 10.1016/j.heliyon.2024.e32965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
The use of animal models for biological experiments is no longer sufficient for research related to human life and disease. The development of organ tissues has replaced animal models by mimicking the structure, function, development and homeostasis of natural organs. This provides more opportunities to study human diseases such as cancer, infectious diseases and genetic disorders. In this study, bibliometric methods were used to analyze organoid-related articles published over the last 20+ years to identify emerging trends and frontiers in organoid research. A total of 13,143 articles from 4125 institutions in 86 countries or regions were included in the analysis. The number of papers increased steadily over the 20-year period. The United States was the leading country in terms of number of papers and citations. Harvard Medical School had the highest number of papers published. Keyword analysis revealed research trends and focus areas such as organ tissues, stem cells, 3D culture and tissue engineering. In conclusion, this study used bibliometric and visualization methods to explore the field of organoid research and found that organ tissues are receiving increasing attention in areas such as cancer, drug discovery, personalized medicine, genetic disease modelling and gene repair, making them a current research hotspot and a future research trend.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jianan Ding
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Zixuan Jia
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guijie Tian
- School of Laboratory Medicine and Biotechnology, Southern Medical University Guangzhou, China
| | - Shuqian Zheng
- School of Basic Medical Sciences, Southern Medical University Guangzhou, China
| | - Pinfei Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jieyan Wang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| | - Hui Liang
- Department of Urology, People's Hospital of Longhua, Shenzhen, Guangdong, 518109, China
| |
Collapse
|
9
|
Manoharan TJM, Ravi K, Suresh AP, Acharya AP, Nikkhah M. Engineered Tumor-Immune Microenvironment On A Chip to Study T Cell-Macrophage Interaction in Breast Cancer Progression. Adv Healthc Mater 2024; 13:e2303658. [PMID: 38358061 PMCID: PMC11146602 DOI: 10.1002/adhm.202303658] [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: 10/23/2023] [Revised: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Evolving knowledge about the tumor-immune microenvironment (TIME) is driving innovation in designing novel therapies against hard-to-treat breast cancer. Targeting the immune components of TIME has emerged as a promising approach for cancer therapy. While recent immunotherapies aim at restoring antitumor immunity, counteracting tumor escape remains challenging. Hence there is a pressing need to better understand the complex tumor-immune crosstalk within TIME. Considering this imperative, this study aims at investigating the crosstalk between the two abundant immune cell populations within the breast TIME-macrophages and T cells, in driving tumor progression using an organotypic 3D in vitro tumor-on-a-chip (TOC) model. The TOC features distinct yet interconnected organotypic tumor and stromal entities. This triculture platform mimics the complex TIME, embedding the two immune populations in a suitable 3D matrix. Analysis of invasion, morphometric measurements, and flow cytometry results underscores the substantial contribution of macrophages to tumor progression, while the presence of T cells is associated with a deceleration in the migratory behavior of both cancer cells and macrophages. Furthermore, cytokine analyses reveal significant upregulation of leptin and RANTES (regulated on activation, normal T Cell expressed and secreted) in triculture. Overall, this study highlights the complexity of TIME and the critical role of immune cells in cancer progression.
Collapse
Affiliation(s)
| | - Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Abhirami P Suresh
- School for Engineering of Matter, Transport and Energy (SEMTE), Arizona State University, Tempe, AZ, 85287, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abhinav P Acharya
- School for Engineering of Matter, Transport and Energy (SEMTE), Arizona State University, Tempe, AZ, 85287, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
- Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA
| |
Collapse
|
10
|
Leal-Alves C, Deng Z, Kermeci N, Shih SCC. Integrating microfluidics and synthetic biology: advancements and diverse applications across organisms. LAB ON A CHIP 2024; 24:2834-2860. [PMID: 38712893 DOI: 10.1039/d3lc01090b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Synthetic biology is the design and modification of biological systems for specific functions, integrating several disciplines like engineering, genetics, and computer science. The field of synthetic biology is to understand biological processes within host organisms through the manipulation and regulation of their genetic pathways and the addition of biocontrol circuits to enhance their production capabilities. This pursuit serves to address global challenges spanning diverse domains that are difficult to tackle through conventional routes of production. Despite its impact, achieving precise, dynamic, and high-throughput manipulation of biological processes is still challenging. Microfluidics offers a solution to those challenges, enabling controlled fluid handling at the microscale, offering lower reagent consumption, faster analysis of biochemical reactions, automation, and high throughput screening. In this review, we diverge from conventional focus on automating the synthetic biology design-build-test-learn cycle, and instead, focus on microfluidic platforms and their role in advancing synthetic biology through its integration with host organisms - bacterial cells, yeast, fungi, animal cells - and cell-free systems. The review illustrates how microfluidic devices have been instrumental in understanding biological systems by showcasing microfluidics as an essential tool to create synthetic genetic circuits, pathways, and organisms within controlled environments. In conclusion, we show how microfluidics expedite synthetic biology applications across diverse domains including but not limited to personalized medicine, bioenergy, and agriculture.
Collapse
Affiliation(s)
- Chiara Leal-Alves
- Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada.
- Department of Electrical and Computer Engineering, Concordia University, 1515 Ste-Catherine St. W, Montréal, QC, H3G1M8 Canada
| | - Zhiyang Deng
- Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada.
- Department of Electrical and Computer Engineering, Concordia University, 1515 Ste-Catherine St. W, Montréal, QC, H3G1M8 Canada
| | - Natalia Kermeci
- Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada.
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada
| | - Steve C C Shih
- Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada.
- Department of Electrical and Computer Engineering, Concordia University, 1515 Ste-Catherine St. W, Montréal, QC, H3G1M8 Canada
- Department of Biology, Concordia University, 7141 Sherbrooke St. W, Montréal, QC, H4B1R6 Canada
| |
Collapse
|
11
|
Veith I, Nurmik M, Mencattini A, Damei I, Lansche C, Brosseau S, Gropplero G, Corgnac S, Filippi J, Poté N, Guenzi E, Chassac A, Mordant P, Tosello J, Sedlik C, Piaggio E, Girard N, Camonis J, Shirvani H, Mami-Chouaib F, Mechta-Grigoriou F, Descroix S, Martinelli E, Zalcman G, Parrini MC. Assessing personalized responses to anti-PD-1 treatment using patient-derived lung tumor-on-chip. Cell Rep Med 2024; 5:101549. [PMID: 38703767 PMCID: PMC11148770 DOI: 10.1016/j.xcrm.2024.101549] [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: 10/28/2023] [Revised: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
There is a compelling need for approaches to predict the efficacy of immunotherapy drugs. Tumor-on-chip technology exploits microfluidics to generate 3D cell co-cultures embedded in hydrogels that recapitulate simplified tumor ecosystems. Here, we present the development and validation of lung tumor-on-chip platforms to quickly and precisely measure ex vivo the effects of immune checkpoint inhibitors on T cell-mediated cancer cell death by exploiting the power of live imaging and advanced image analysis algorithms. The integration of autologous immunosuppressive FAP+ cancer-associated fibroblasts impaired the response to anti-PD-1, indicating that tumors-on-chips are capable of recapitulating stroma-dependent mechanisms of immunotherapy resistance. For a small cohort of non-small cell lung cancer patients, we generated personalized tumors-on-chips with their autologous primary cells isolated from fresh tumor samples, and we measured the responses to anti-PD-1 treatment. These results support the power of tumor-on-chip technology in immuno-oncology research and open a path to future clinical validations.
Collapse
Affiliation(s)
- Irina Veith
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France; Institut Roche, 30 Cours de l'Île Seguin, 92100 Boulogne-Billancourt, France
| | - Martin Nurmik
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France
| | - Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Isabelle Damei
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Christine Lansche
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France
| | - Solenn Brosseau
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France; Université Paris Cité, Thoracic Oncology Department and CIC INSERM 1425, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Giacomo Gropplero
- Institut Curie, CNRS UMR168, Laboratoire Physico Chimie Curie, Institut Pierre-Gilles de Gennes, PSL Research University, 75005 Paris, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Joanna Filippi
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Nicolas Poté
- Université Paris Cité, INSERM UMR1152, Hôpital Bichat-Claude Bernard, 75018 Paris, France; Department of Pathology, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Edouard Guenzi
- Université Paris Cité, INSERM UMR1152, Hôpital Bichat-Claude Bernard, 75018 Paris, France; Department of Pathology, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Anaïs Chassac
- Department of Pathology, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Pierre Mordant
- Université Paris Cité, Thoracic Surgery Department, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Jimena Tosello
- INSERM U932, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Christine Sedlik
- INSERM U932, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Eliane Piaggio
- INSERM U932, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, PSL Research University, Institut Curie Research Center, Paris, France
| | - Nicolas Girard
- INSERM U932, PSL Research University, Institut Curie Research Center, Paris, France; Institut Curie, Institut du Thorax Curie Montsouris, Paris, France; Paris Saclay University, UVSQ, Versailles, France
| | - Jacques Camonis
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France
| | - Hamasseh Shirvani
- Institut Roche, 30 Cours de l'Île Seguin, 92100 Boulogne-Billancourt, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Université Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Fatima Mechta-Grigoriou
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France
| | - Stéphanie Descroix
- Institut Curie, CNRS UMR168, Laboratoire Physico Chimie Curie, Institut Pierre-Gilles de Gennes, PSL Research University, 75005 Paris, France
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gérard Zalcman
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France; Université Paris Cité, Thoracic Oncology Department and CIC INSERM 1425, Hôpital Bichat-Claude Bernard, 75018 Paris, France.
| | - Maria Carla Parrini
- Institut Curie, INSERM U830, Stress and Cancer Laboratory, PSL Research University, 26 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
12
|
Skirzynska A, Xue C, Shoichet MS. Engineering Biomaterials to Model Immune-Tumor Interactions In Vitro. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310637. [PMID: 38349174 DOI: 10.1002/adma.202310637] [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: 10/12/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Engineered biomaterial scaffolds are becoming more prominent in research laboratories to study drug efficacy for oncological applications in vitro, but do they have a place in pharmaceutical drug screening pipelines? The low efficacy of cancer drugs in phase II/III clinical trials suggests that there are critical mechanisms not properly accounted for in the pre-clinical evaluation of drug candidates. Immune cells associated with the tumor may account for some of these failures given recent successes with cancer immunotherapies; however, there are few representative platforms to study immune cells in the context of cancer as traditional 2D culture is typically monocultures and humanized animal models have a weakened immune composition. Biomaterials that replicate tumor microenvironmental cues may provide a more relevant model with greater in vitro complexity. In this review, the authors explore the pertinent microenvironmental cues that drive tumor progression in the context of the immune system, discuss how these cues can be incorporated into hydrogel design to culture immune cells, and describe progress toward precision oncological drug screening with engineered tissues.
Collapse
Affiliation(s)
- Arianna Skirzynska
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
| | - Chang Xue
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
| | - Molly S Shoichet
- Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, 160 College St, Toronto, ON, M5S 3E1, Canada
- Institute for Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 College Street, Toronto, ON, M5S 3H4, Canada
| |
Collapse
|
13
|
Ghoshal D, Petersen I, Ringquist R, Kramer L, Bhatia E, Hu T, Richard A, Park R, Corbin J, Agarwal S, Thomas A, Ramirez S, Tharayil J, Downey E, Ketchum F, Ochal A, Sonthi N, Lonial S, Kochenderfer JN, Tran R, Zhu M, Lam WA, Coskun AF, Roy K. Multi-Niche Human Bone Marrow On-A-Chip for Studying the Interactions of Adoptive CAR-T Cell Therapies with Multiple Myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588601. [PMID: 38644993 PMCID: PMC11030357 DOI: 10.1101/2024.04.08.588601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Multiple myeloma (MM), a cancer of bone marrow plasma cells, is the second-most common hematological malignancy. However, despite immunotherapies like chimeric antigen receptor (CAR)-T cells, relapse is nearly universal. The bone marrow (BM) microenvironment influences how MM cells survive, proliferate, and resist treatment. Yet, it is unclear which BM niches give rise to MM pathophysiology. Here, we present a 3D microvascularized culture system, which models the endosteal and perivascular bone marrow niches, allowing us to study MM-stroma interactions in the BM niche and model responses to therapeutic CAR-T cells. We demonstrated the prolonged survival of cell line-based and patient-derived multiple myeloma cells within our in vitro system and successfully flowed in donor-matched CAR-T cells. We then measured T cell survival, differentiation, and cytotoxicity against MM cells using a variety of analysis techniques. Our MM-on-a-chip system could elucidate the role of the BM microenvironment in MM survival and therapeutic evasion and inform the rational design of next-generation therapeutics. TEASER A multiple myeloma model can study why the disease is still challenging to treat despite options that work well in other cancers.
Collapse
|
14
|
Bonnet V, Maikranz E, Madec M, Vertti-Quintero N, Cuche C, Mastrogiovanni M, Alcover A, Di Bartolo V, Baroud CN. Cancer-on-a-chip model shows that the adenomatous polyposis coli mutation impairs T cell engagement and killing of cancer spheroids. Proc Natl Acad Sci U S A 2024; 121:e2316500121. [PMID: 38442157 PMCID: PMC10945811 DOI: 10.1073/pnas.2316500121] [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: 09/22/2023] [Accepted: 01/25/2024] [Indexed: 03/07/2024] Open
Abstract
Evaluating the ability of cytotoxic T lymphocytes (CTLs) to eliminate tumor cells is crucial, for instance, to predict the efficiency of cell therapy in personalized medicine. However, the destruction of a tumor by CTLs involves CTL migration in the extra-tumoral environment, accumulation on the tumor, antigen recognition, and cooperation in killing the cancer cells. Therefore, identifying the limiting steps in this complex process requires spatio-temporal measurements of different cellular events over long periods. Here, we use a cancer-on-a-chip platform to evaluate the impact of adenomatous polyposis coli (APC) mutation on CTL migration and cytotoxicity against 3D tumor spheroids. The APC mutated CTLs are found to have a reduced ability to destroy tumor spheroids compared with control cells, even though APC mutants migrate in the extra-tumoral space and accumulate on the spheroids as efficiently as control cells. Once in contact with the tumor however, mutated CTLs display reduced engagement with the cancer cells, as measured by a metric that distinguishes different modes of CTL migration. Realigning the CTL trajectories around localized killing cascades reveals that all CTLs transition to high engagement in the 2 h preceding the cascades, which confirms that the low engagement is the cause of reduced cytotoxicity. Beyond the study of APC mutations, this platform offers a robust way to compare cytotoxic cell efficiency of even closely related cell types, by relying on a multiscale cytometry approach to disentangle complex interactions and to identify the steps that limit the tumor destruction.
Collapse
Affiliation(s)
- Valentin Bonnet
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Erik Maikranz
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Marianne Madec
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Faculty of Medicine, Department of Pathology and Immunology, University of Geneva, Geneva 4CH-1211, Switzerland
| | - Nadia Vertti-Quintero
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
| | - Céline Cuche
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Marta Mastrogiovanni
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Albert Einstein College of Medicine, Department of Developmental and Molecular Biology, New York, NY10461
| | - Andrés Alcover
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Vincenzo Di Bartolo
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Charles N. Baroud
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| |
Collapse
|
15
|
Ravi K, Manoharan TJM, Wang KC, Pockaj B, Nikkhah M. Engineered 3D ex vivo models to recapitulate the complex stromal and immune interactions within the tumor microenvironment. Biomaterials 2024; 305:122428. [PMID: 38147743 PMCID: PMC11098715 DOI: 10.1016/j.biomaterials.2023.122428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
Cancer thrives in a complex environment where interactions between cellular and acellular components, surrounding the tumor, play a crucial role in disease development and progression. Despite significant progress in cancer research, the mechanism driving tumor growth and therapeutic outcomes remains elusive. Two-dimensional (2D) cell culture assays and in vivo animal models are commonly used in cancer research and therapeutic testing. However, these models suffer from numerous shortcomings including lack of key features of the tumor microenvironment (TME) & cellular composition, cost, and ethical clearance. To that end, there is an increased interest in incorporating and elucidating the influence of TME on cancer progression. Advancements in 3D-engineered ex vivo models, leveraging biomaterials and microengineering technologies, have provided an unprecedented ability to reconstruct native-like bioengineered cancer models to study the heterotypic interactions of TME with a spatiotemporal organization. These bioengineered cancer models have shown excellent capabilities to bridge the gap between oversimplified 2D systems and animal models. In this review article, we primarily provide an overview of the immune and stromal cellular components of the TME and then discuss the latest state-of-the-art 3D-engineered ex vivo platforms aiming to recapitulate the complex TME features. The engineered TME model, discussed herein, are categorized into three main sections according to the cellular interactions within TME: (i) Tumor-Stromal interactions, (ii) Tumor-Immune interactions, and (iii) Complex TME interactions. Finally, we will conclude the article with a perspective on how these models can be instrumental for cancer translational studies and therapeutic testing.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
16
|
Tostado CP, Da Ong LX, Heng JJW, Miccolis C, Chia S, Seow JJW, Toh Y, DasGupta R. An AI-assisted integrated, scalable, single-cell phenomic-transcriptomic platform to elucidate intratumor heterogeneity against immune response. Bioeng Transl Med 2024; 9:e10628. [PMID: 38435825 PMCID: PMC10905538 DOI: 10.1002/btm2.10628] [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: 11/07/2023] [Accepted: 11/16/2023] [Indexed: 03/05/2024] Open
Abstract
We present a novel framework combining single-cell phenotypic data with single-cell transcriptomic analysis to identify factors underpinning heterogeneity in antitumor immune response. We developed a pairwise, tumor-immune discretized interaction assay between natural killer (NK-92MI) cells and patient-derived head and neck squamous cell carcinoma (HNSCC) cell lines on a microfluidic cell-trapping platform. Furthermore we generated a deep-learning computer vision algorithm that is capable of automating the acquisition and analysis of a large, live-cell imaging data set (>1 million) of paired tumor-immune interactions spanning a time course of 24 h across multiple HNSCC lines (n = 10). Finally, we combined the response data measured by Kaplan-Meier survival analysis against NK-mediated killing with downstream single-cell transcriptomic analysis to interrogate molecular signatures associated with NK-effector response. As proof-of-concept for the proposed framework, we efficiently identified MHC class I-driven cytotoxic resistance as a key mechanism for immune evasion in nonresponders, while enhanced expression of cell adhesion molecules was found to be correlated with sensitivity against NK-mediated cytotoxicity. We conclude that this integrated, data-driven phenotypic approach holds tremendous promise in advancing the rapid identification of new mechanisms and therapeutic targets related to immune evasion and response.
Collapse
Affiliation(s)
- Christopher P. Tostado
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
- Institute for Health Innovation and Technology (iHealthtech), National University of SingaporeSingaporeSingapore
| | - Lucas Xian Da Ong
- Institute for Health Innovation and Technology (iHealthtech), National University of SingaporeSingaporeSingapore
| | - Joel Jia Wei Heng
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
| | - Carlo Miccolis
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
| | - Shumei Chia
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
| | - Justine Jia Wen Seow
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
| | - Yi‐Chin Toh
- Institute for Health Innovation and Technology (iHealthtech), National University of SingaporeSingaporeSingapore
- School of Mechanical, Medical and Process EngineeringQueensland University of TechnologyBrisbaneAustralia
- Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneAustralia
| | - Ramanuj DasGupta
- Genome Institute of Singapore, Laboratory of Precision Oncology and Cancer EvolutionSingaporeSingapore
| |
Collapse
|
17
|
Wu Z, Huang D, Wang J, Zhao Y, Sun W, Shen X. Engineering Heterogeneous Tumor Models for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304160. [PMID: 37946674 PMCID: PMC10767453 DOI: 10.1002/advs.202304160] [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: 06/22/2023] [Revised: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Tumor tissue engineering holds great promise for replicating the physiological and behavioral characteristics of tumors in vitro. Advances in this field have led to new opportunities for studying the tumor microenvironment and exploring potential anti-cancer therapeutics. However, the main obstacle to the widespread adoption of tumor models is the poor understanding and insufficient reconstruction of tumor heterogeneity. In this review, the current progress of engineering heterogeneous tumor models is discussed. First, the major components of tumor heterogeneity are summarized, which encompasses various signaling pathways, cell proliferations, and spatial configurations. Then, contemporary approaches are elucidated in tumor engineering that are guided by fundamental principles of tumor biology, and the potential of a bottom-up approach in tumor engineering is highlighted. Additionally, the characterization approaches and biomedical applications of tumor models are discussed, emphasizing the significant role of engineered tumor models in scientific research and clinical trials. Lastly, the challenges of heterogeneous tumor models in promoting oncology research and tumor therapy are described and key directions for future research are provided.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Jinglin Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
| | - Xian Shen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325035China
| |
Collapse
|
18
|
Abizanda-Campo S, Virumbrales-Muñoz M, Humayun M, Marmol I, Beebe DJ, Ochoa I, Oliván S, Ayuso JM. Microphysiological systems for solid tumor immunotherapy: opportunities and challenges. MICROSYSTEMS & NANOENGINEERING 2023; 9:154. [PMID: 38106674 PMCID: PMC10724276 DOI: 10.1038/s41378-023-00616-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023]
Abstract
Immunotherapy remains more effective for hematologic tumors than for solid tumors. One of the main challenges to immunotherapy of solid tumors is the immunosuppressive microenvironment these tumors generate, which limits the cytotoxic capabilities of immune effector cells (e.g., cytotoxic T and natural killer cells). This microenvironment is characterized by hypoxia, nutrient starvation, accumulated waste products, and acidic pH. Tumor-hijacked cells, such as fibroblasts, macrophages, and T regulatory cells, also contribute to this inhospitable microenvironment for immune cells by secreting immunosuppressive cytokines that suppress the antitumor immune response and lead to immune evasion. Thus, there is a strong interest in developing new drugs and cell formulations that modulate the tumor microenvironment and reduce tumor cell immune evasion. Microphysiological systems (MPSs) are versatile tools that may accelerate the development and evaluation of these therapies, although specific examples showcasing the potential of MPSs remain rare. Advances in microtechnologies have led to the development of sophisticated microfluidic devices used to recapitulate tumor complexity. The resulting models, also known as microphysiological systems (MPSs), are versatile tools with which to decipher the molecular mechanisms driving immune cell antitumor cytotoxicity, immune cell exhaustion, and immune cell exclusion and to evaluate new targeted immunotherapies. Here, we review existing microphysiological platforms to study immuno-oncological applications and discuss challenges and opportunities in the field.
Collapse
Affiliation(s)
- Sara Abizanda-Campo
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI USA
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - María Virumbrales-Muñoz
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI USA
| | - Mouhita Humayun
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Cambridge, MA USA
| | - Ines Marmol
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI USA
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Sara Oliván
- Tissue Microenvironment Lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, Spain
| | - Jose M Ayuso
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI USA
- University of Wisconsin Carbone Cancer Center, Madison, WI USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI USA
| |
Collapse
|
19
|
Yu Y, Zhou T, Cao L. Use and application of organ-on-a-chip platforms in cancer research. J Cell Commun Signal 2023:10.1007/s12079-023-00790-7. [PMID: 38032444 DOI: 10.1007/s12079-023-00790-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Tumors are a major cause of death worldwide, and much effort has been made to develop appropriate anti-tumor therapies. Existing in vitro and in vivo tumor models cannot reflect the critical features of cancer. The development of organ-on-a-chip models has enabled the integration of organoids, microfluidics, tissue engineering, biomaterials research, and microfabrication, offering conditions that mimic tumor physiology. Three-dimensional in vitro human tumor models that have been established as organ-on-a-chip models contain multiple cell types and a structure that is similar to the primary tumor. These models can be applied to various foci of oncology research. Moreover, the high-throughput features of microfluidic organ-on-a-chip models offer new opportunities for achieving large-scale drug screening and developing more personalized treatments. In this review of the literature, we explore the development of organ-on-a-chip technology and discuss its use as an innovative tool in basic and clinical applications and summarize its advancement of cancer research.
Collapse
Affiliation(s)
- Yifan Yu
- Department of Hepatobiliary and Transplant Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - TingTing Zhou
- The College of Basic Medical Science, Health Sciences Institute, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Liu Cao
- The College of Basic Medical Science, Health Sciences Institute, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
20
|
Wirthl B, Janko C, Lyer S, Schrefler BA, Alexiou C, Wall WA. An in silico model of the capturing of magnetic nanoparticles in tumour spheroids in the presence of flow. Biomed Microdevices 2023; 26:1. [PMID: 38008813 PMCID: PMC10678808 DOI: 10.1007/s10544-023-00685-9] [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] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
One of the main challenges in improving the efficacy of conventional chemotherapeutic drugs is that they do not reach the cancer cells at sufficiently high doses while at the same time affecting healthy tissue and causing significant side effects and suffering in cancer patients. To overcome this deficiency, magnetic nanoparticles as transporter systems have emerged as a promising approach to achieve more specific tumour targeting. Drug-loaded magnetic nanoparticles can be directed to the target tissue by applying an external magnetic field. However, the magnetic forces exerted on the nanoparticles fall off rapidly with distance, making the tumour targeting challenging, even more so in the presence of flowing blood or interstitial fluid. We therefore present a computational model of the capturing of magnetic nanoparticles in a test setup: our model includes the flow around the tumour, the magnetic forces that guide the nanoparticles, and the transport within the tumour. We show how a model for the transport of magnetic nanoparticles in an external magnetic field can be integrated with a multiphase tumour model based on the theory of porous media. Our approach based on the underlying physical mechanisms can provide crucial insights into mechanisms that cannot be studied conclusively in experimental research alone. Such a computational model enables an efficient and systematic exploration of the nanoparticle design space, first in a controlled test setup and then in more complex in vivo scenarios. As an effective tool for minimising costly trial-and-error design methods, it expedites translation into clinical practice to improve therapeutic outcomes and limit adverse effects for cancer patients.
Collapse
Affiliation(s)
- Barbara Wirthl
- Institute for Computational Mechanics, Technical University of Munich, TUM School of Engineering and Design, Department of Engineering Physics & Computation, Garching bei München, Germany.
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Guided Nanomaterials within the framework of the Hightech Agenda (HTA) of the Free State of Bavaria, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Bernhard A Schrefler
- Department of Civil, Environmental and Architectural Engineering, University of Padua, Padua, Italy
- Institute for Advanced Study, Technical University of Munich, Garching bei München, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolfgang A Wall
- Institute for Computational Mechanics, Technical University of Munich, TUM School of Engineering and Design, Department of Engineering Physics & Computation, Garching bei München, Germany
| |
Collapse
|
21
|
Ayensa-Jiménez J, Doweidar MH, Doblaré M, Gaffney EA. A Mathematical Modelling Study of Chemotactic Dynamics in Cell Cultures: The Impact of Spatio-temporal Heterogeneity. Bull Math Biol 2023; 85:98. [PMID: 37684435 PMCID: PMC10491576 DOI: 10.1007/s11538-023-01194-9] [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: 12/19/2022] [Accepted: 08/04/2023] [Indexed: 09/10/2023]
Abstract
As motivated by studies of cellular motility driven by spatiotemporal chemotactic gradients in microdevices, we develop a framework for constructing approximate analytical solutions for the location, speed and cellular densities for cell chemotaxis waves in heterogeneous fields of chemoattractant from the underlying partial differential equation models. In particular, such chemotactic waves are not in general translationally invariant travelling waves, but possess a spatial variation that evolves in time, and may even oscillate back and forth in time, according to the details of the chemotactic gradients. The analytical framework exploits the observation that unbiased cellular diffusive flux is typically small compared to chemotactic fluxes and is first developed and validated for a range of exemplar scenarios. The framework is subsequently applied to more complex models considering the chemoattractant dynamics under more general settings, potentially including those of relevance for representing pathophysiology scenarios in microdevice studies. In particular, even though solutions cannot be constructed in all cases, a wide variety of scenarios can be considered analytically, firstly providing global insight into the important mechanisms and features of cell motility in complex spatiotemporal fields of chemoattractant. Such analytical solutions also provide a means of rapid evaluation of model predictions, with the prospect of application in computationally demanding investigations relating theoretical models and experimental observation, such as Bayesian parameter estimation.
Collapse
Affiliation(s)
- Jacobo Ayensa-Jiménez
- Aragon Institute of Engineering Research, University of Zaragoza, Mariano Esquillor, s.n., 50018 Zaragoza, Spain
- Tissue Microenvironment Laboratory (TME Lab), Institute for Health Research Aragón, San Juan Bosco, 13, 50009 Zaragoza, Spain
| | - Mohamed H. Doweidar
- Aragon Institute of Engineering Research, University of Zaragoza, Mariano Esquillor, s.n., 50018 Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, María de Luna s.n., 50018 Zaragoza, Spain
| | - Manuel Doblaré
- Aragon Institute of Engineering Research, University of Zaragoza, Mariano Esquillor, s.n., 50018 Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, María de Luna s.n., 50018 Zaragoza, Spain
- Tissue Microenvironment Laboratory (TME Lab), Institute for Health Research Aragón, San Juan Bosco, 13, 50009 Zaragoza, Spain
- Nanjing Tech University, 30 South Puzhu Road, 211816 Nanjing, China
| | - Eamonn A. Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Woodstock Road, Oxford, OX2 6GG UK
| |
Collapse
|
22
|
Yang Q, Li M, Yang X, Xiao Z, Tong X, Tuerdi A, Li S, Lei L. Flourishing tumor organoids: History, emerging technology, and application. Bioeng Transl Med 2023; 8:e10559. [PMID: 37693042 PMCID: PMC10487342 DOI: 10.1002/btm2.10559] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/16/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Malignant tumors are one of the leading causes of death which impose an increasingly heavy burden on all countries. Therefore, the establishment of research models that closely resemble original tumor characteristics is crucial to further understanding the mechanisms of malignant tumor development, developing safer and more effective drugs, and formulating personalized treatment plans. Recently, organoids have been widely used in tumor research owing to their advantages including preserving the structure, heterogeneity, and cellular functions of the original tumor, together with the ease of manipulation. This review describes the history and characteristics of tumor organoids and the synergistic combination of three-dimensional (3D) culture approaches for tumor organoids with emerging technologies, including tissue-engineered cell scaffolds, microfluidic devices, 3D bioprinting, rotating wall vessels, and clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR-Cas9). Additionally, the progress in research and the applications in basic and clinical research of tumor organoid models are summarized. This includes studies of the mechanism of tumor development, drug development and screening, precision medicine, immunotherapy, and simulation of the tumor microenvironment. Finally, the existing shortcomings of tumor organoids and possible future directions are discussed.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ayinuer Tuerdi
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
23
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
24
|
Barozzi D, Scielzo C. Emerging Strategies in 3D Culture Models for Hematological Cancers. Hemasphere 2023; 7:e932. [PMID: 37520775 PMCID: PMC10378728 DOI: 10.1097/hs9.0000000000000932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023] Open
Abstract
In vitro cell cultures are fundamental and necessary tools in cancer research and personalized drug discovery. Currently, most cells are cultured using two-dimensional (2D) methods, and drug testing is mainly performed in animal models. However, new and improved methods that implement three-dimensional (3D) cell-culturing techniques provide compelling evidence that more advanced experiments can be performed, yielding valuable new insights. In 3D cell-culture experiments, the cell environment can be manipulated to mimic the complexity and dynamicity of the human tissue microenvironment, possibly leading to more accurate representations of cell-to-cell interactions, tumor biology, and predictions of drug response. The 3D cell cultures can also potentially provide alternative ways to study hematological cancers and are expected to eventually bridge the gap between 2D cell culture and animal models. The present review provides an overview of the complexity of the lymphoid microenvironment and a summary of the currently used 3D models that aim at recreating it for hematological cancer research. We here dissect the differences and challenges between, and potential advantages of, different culture methods and present our vision of the most promising future strategies in the hematological field.
Collapse
Affiliation(s)
- Dafne Barozzi
- Università degli Studi di Milano-Bicocca, School of Medicine and Surgery, PhD program in Molecular and Translational Medicine (DIMET), Milano, Italy
- Unit of Malignant B cells biology and 3D modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Cristina Scielzo
- Unit of Malignant B cells biology and 3D modelling, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milano, Italy
| |
Collapse
|
25
|
Salehi A, Naserzadeh P, Tarighi P, Afjeh-Dana E, Akhshik M, Jafari A, Mackvandi P, Ashtari B, Mozafari M. Fabrication of a microfluidic device for probiotic drug's dosage screening: Precision Medicine for Breast Cancer Treatment. Transl Oncol 2023; 34:101674. [PMID: 37224765 PMCID: PMC10302160 DOI: 10.1016/j.tranon.2023.101674] [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: 09/14/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 05/26/2023] Open
Abstract
Breast cancer is the most common cancer in women; it has been affecting the lives of millions each year globally and microfluidic devices seem to be a promising method for the future advancements in this field. This research uses a dynamic cell culture condition in a microfluidic concentration gradient device, helping us to assess breast anticancer activities of probiotic strains against MCF-7 cells. It has been shown that MCF-7 cells could grow and proliferate for at least 24 h; however, a specific concentration of probiotic supernatant could induce more cell death signaling population after 48 h. One of our key findings was that our evaluated optimum dose (7.8 mg/L) was less than the conventional static cell culture treatment dose (12 mg/L). To determine the most effective dose over time and the percentage of apoptosis versus necrosis, flowcytometric assessment was performed. Exposing the MCF-7 cells to probiotic supernatant after 6, 24 and 48 h, confirmed that the apoptotic and necrotic cell death signaling were concentration and time dependent. We have shown a case that these types of microfluidics platforms performing dynamic cell culture could be beneficial in personalized medicine and cancer therapy.
Collapse
Affiliation(s)
- Ali Salehi
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences. Tehran, Iran
| | - Parvaneh Naserzadeh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences. Tehran, Iran
| | - Elham Afjeh-Dana
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Akhshik
- Centre for Biocomposites and Biomaterials Processing. University of Toronto, Canada; EPICentre, University of Windsor, Canada
| | - Amir Jafari
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences. Tehran, Iran
| | - Pooyan Mackvandi
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences. Tehran, Iran; Centre for Materials Interfaces, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, Pontedera, 56025 Pisa, Italy
| | - Behnaz Ashtari
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences. Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Masoud Mozafari
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
26
|
Engrácia DM, Pinto CIG, Mendes F. Cancer 3D Models for Metallodrug Preclinical Testing. Int J Mol Sci 2023; 24:11915. [PMID: 37569291 PMCID: PMC10418685 DOI: 10.3390/ijms241511915] [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: 06/04/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Despite being standard tools in research, the application of cellular and animal models in drug development is hindered by several limitations, such as limited translational significance, animal ethics, and inter-species physiological differences. In this regard, 3D cellular models can be presented as a step forward in biomedical research, allowing for mimicking tissue complexity more accurately than traditional 2D models, while also contributing to reducing the use of animal models. In cancer research, 3D models have the potential to replicate the tumor microenvironment, which is a key modulator of cancer cell behavior and drug response. These features make cancer 3D models prime tools for the preclinical study of anti-tumoral drugs, especially considering that there is still a need to develop effective anti-cancer drugs with high selectivity, minimal toxicity, and reduced side effects. Metallodrugs, especially transition-metal-based complexes, have been extensively studied for their therapeutic potential in cancer therapy due to their distinctive properties; however, despite the benefits of 3D models, their application in metallodrug testing is currently limited. Thus, this article reviews some of the most common types of 3D models in cancer research, as well as the application of 3D models in metallodrug preclinical studies.
Collapse
Affiliation(s)
- Diogo M. Engrácia
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Catarina I. G. Pinto
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Filipa Mendes
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
- Department of Nuclear Sciences and Engineering, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
27
|
Beelen NA, Aberle MR, Bruno V, Olde Damink SWM, Bos GMJ, Rensen SS, Wieten L. Antibody-dependent cellular cytotoxicity-inducing antibodies enhance the natural killer cell anti-cancer response against patient-derived pancreatic cancer organoids. Front Immunol 2023; 14:1133796. [PMID: 37520563 PMCID: PMC10375290 DOI: 10.3389/fimmu.2023.1133796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/13/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Pancreatic cancer is associated with poor prognosis, and limited treatment options are available for the majority of patients. Natural killer (NK) cells in combination with antibodies inducing antibody-dependent cell-mediated cytotoxicity (ADCC) could be a highly effective new therapeutic option in pancreatic cancer. Accurate predictive preclinical models are needed to develop successful NK cell immunotherapy. Tumor organoids, in vitro 3D organ-like structures that retain important pathophysiological characteristics of the in vivo tumor, may provide such a model. In the current study, we assessed the cytotoxic potential of adoptive NK cells against human pancreatic cancer organoids. We hypothesized that NK cell anti-tumor responses could be enhanced by including ADCC-triggering antibodies. Methods We performed cytotoxicity assays with healthy donor-derived IL-2-activated NK cells and pancreatic cancer organoids from four patients. A 3D cytotoxicity assay using live-cell-imaging was developed and enabled real-time assessment of the response. Results We show that NK cells migrate to and target pancreatic cancer organoids, resulting in an increased organoid death, compared to the no NK cell controls (reaching an average fold change from baseline of 2.1±0.8 vs 1.4±0.6). After 24-hours of co-culture, organoid 2D growth increased. Organoids from 2 out of 4 patients were sensitive to NK cells, while organoids from the other two patients were relatively resistant, indicating patient-specific heterogeneity among organoid cultures. The ADCC-inducing antibodies avelumab (anti-PD-L1) and trastuzumab (anti-HER2) increased NK cell-induced organoid cell death (reaching an average fold change from baseline of 3.5±1.0 and 4.5±1.8, respectively). Moreover, combination therapy with avelumab or trastuzumab resulted in complete disintegration of organoids. Finally, inclusion of ADCC-inducing antibodies was able to overcome resistance in NK-organoid combinations with low or no kill. Discussion These results support the use of organoids as a relevant and personalized model to study the anti-tumor response of NK cells in vitro and the potential of ADCC-inducing antibodies to enhance NK cell effector function.
Collapse
Affiliation(s)
- Nicky A. Beelen
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Merel R. Aberle
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
- Department of Pharmacology and Toxicology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Virginia Bruno
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Steven W. M. Olde Damink
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
- Department of General, Visceral- and Transplantation Surgery, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Gerard M. J. Bos
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, Netherlands
- GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Sander S. Rensen
- Department of Surgery and School of Nutrition and Translational Research in Metabolism (NUTRIM), School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Lotte Wieten
- GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
28
|
González-Lana S, Randelovic T, Ciriza J, López-Valdeolivas M, Monge R, Sánchez-Somolinos C, Ochoa I. Surface modifications of COP-based microfluidic devices for improved immobilisation of hydrogel proteins: long-term 3D culture with contractile cell types and ischaemia model. LAB ON A CHIP 2023; 23:2434-2446. [PMID: 37013698 DOI: 10.1039/d3lc00075c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The tissue microenvironment plays a crucial role in tissue homeostasis and disease progression. However, the in vitro simulation has been limited by the lack of adequate biomimetic models in the last decades. Thanks to the advent of microfluidic technology for cell culture applications, these complex microenvironments can be recreated by combining hydrogels, cells and microfluidic devices. Nevertheless, this advance has several limitations. When cultured in three-dimensional (3D) hydrogels inside microfluidic devices, contractile cells may exert forces that eventually collapse the 3D structure. Disrupting the compartmentalisation creates an obstacle to long-term or highly cell-concentrated assays, which are extremely relevant for multiple applications such as fibrosis or ischaemia. Therefore, we tested surface treatments on cyclic-olefin polymer-based microfluidic devices (COP-MD) to promote the immobilisation of collagen as a 3D matrix protein. Thus, we compared three surface treatments in COP devices for culturing human cardiac fibroblasts (HCF) embedded in collagen hydrogels. We determined the immobilisation efficiency of collagen hydrogel by quantifying the hydrogel transversal area within the devices at the studied time points. Altogether, our results indicated that surface modification with polyacrylic acid photografting (PAA-PG) of COP-MD is the most effective treatment to avoid the quick collapse of collagen hydrogels. As a proof-of-concept experiment, and taking advantage of the low-gas permeability properties of COP-MD, we studied the application of PAA-PG pre-treatment to generate a self-induced ischaemia model. Different necrotic core sizes were developed depending on initial HCF density seeding with no noticeable gel collapse. We conclude that PAA-PG allows long-term culture, gradient generation and necrotic core formation of contractile cell types such as myofibroblasts. This novel approach will pave the way for new relevant in vitro co-culture models where fibroblasts play a key role such as wound healing, tumour microenvironment and ischaemia within microfluidic devices.
Collapse
Affiliation(s)
- Sandra González-Lana
- Tissue Microenvironment (TME) Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, C/ Mariano Esquillor s/n, 500018 Zaragoza, Spain.
- BEONCHIP S.L., CEMINEM, Campus Río Ebro. C/ Mariano Esquillor Gómez s/n, 50018 Zaragoza, Spain
| | - Teodora Randelovic
- Tissue Microenvironment (TME) Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, C/ Mariano Esquillor s/n, 500018 Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Paseo de Isabel La Católica 1-3, 50009 Zaragoza, Spain
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Jesús Ciriza
- Tissue Microenvironment (TME) Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, C/ Mariano Esquillor s/n, 500018 Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Paseo de Isabel La Católica 1-3, 50009 Zaragoza, Spain
| | - María López-Valdeolivas
- Aragón Institute of Nanoscience and Materials (INMA), Department of Condensed Matter Physics (Faculty of Science), CSIC-University of Zaragoza, C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Rosa Monge
- BEONCHIP S.L., CEMINEM, Campus Río Ebro. C/ Mariano Esquillor Gómez s/n, 50018 Zaragoza, Spain
| | - Carlos Sánchez-Somolinos
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Aragón Institute of Nanoscience and Materials (INMA), Department of Condensed Matter Physics (Faculty of Science), CSIC-University of Zaragoza, C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, C/ Mariano Esquillor s/n, 500018 Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Paseo de Isabel La Católica 1-3, 50009 Zaragoza, Spain
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
29
|
Manduca N, Maccafeo E, De Maria R, Sistigu A, Musella M. 3D cancer models: One step closer to in vitro human studies. Front Immunol 2023; 14:1175503. [PMID: 37114038 PMCID: PMC10126361 DOI: 10.3389/fimmu.2023.1175503] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer immunotherapy is the great breakthrough in cancer treatment as it displayed prolonged progression-free survival over conventional therapies, yet, to date, in only a minority of patients. In order to broad cancer immunotherapy clinical applicability some roadblocks need to be overcome, first among all the lack of preclinical models that faithfully depict the local tumor microenvironment (TME), which is known to dramatically affect disease onset, progression and response to therapy. In this review, we provide the reader with a detailed overview of current 3D models developed to mimick the complexity and the dynamics of the TME, with a focus on understanding why the TME is a major target in anticancer therapy. We highlight the advantages and translational potentials of tumor spheroids, organoids and immune Tumor-on-a-Chip models in disease modeling and therapeutic response, while outlining pending challenges and limitations. Thinking forward, we focus on the possibility to integrate the know-hows of micro-engineers, cancer immunologists, pharmaceutical researchers and bioinformaticians to meet the needs of cancer researchers and clinicians interested in using these platforms with high fidelity for patient-tailored disease modeling and drug discovery.
Collapse
Affiliation(s)
- Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ‘A. Gemelli’ - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
30
|
Fan Y, Xu C, Deng N, Gao Z, Jiang Z, Li X, Zhou Y, Pei H, Li L, Tang B. Understanding drug nanocarrier and blood-brain barrier interaction based on a microfluidic microphysiological model. LAB ON A CHIP 2023; 23:1935-1944. [PMID: 36891748 DOI: 10.1039/d2lc01077a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
As many nanoparticles (NPs) have been exploited as drug carriers to overcome the resistance of the blood-brain barrier (BBB), reliable in vitro BBB models are urgently needed to help researchers to comprehensively understand drug nanocarrier-BBB interaction during penetration, which can prompt pre-clinical nanodrug exploitation. Herein, we developed a microfluidic microphysiological model, allowing the analysis of BBB homeostasis and NP penetration. We found that the BBB penetrability of gold nanoparticles (AuNPs) was size- and modification-dependent, which might be caused by a distinct transendocytosis pathway. Notably, transferrin-modified 13 nm AuNPs held the strongest BBB penetrability and induced the slightest BBB dysfunction, while bare 80 nm and 120 nm AuNPs showed opposite results. Moreover, further analysis of the protein corona showed that PEGylation reduced the protein absorption, and some proteins facilitated the BBB penetration of NPs. The developed microphysiological model provides a powerful tool for understanding the drug nanocarrier-BBB interaction, which is vital for exploiting high-efficiency and biocompatible nanodrugs.
Collapse
Affiliation(s)
- Yuanyuan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Chang Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Ning Deng
- Shandong Institute for Product Quality Inspection, Jinan 250101, P. R. China
| | - Ze Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Zhongyao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Xiaoxiao Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Yingshun Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
31
|
Dennison R, Usuga E, Chen H, Paul JZ, Arbelaez CA, Teng YD. Direct Cell Reprogramming and Phenotypic Conversion: An Analysis of Experimental Attempts to Transform Astrocytes into Neurons in Adult Animals. Cells 2023; 12:618. [PMID: 36831283 PMCID: PMC9954435 DOI: 10.3390/cells12040618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Central nervous system (CNS) repair after injury or disease remains an unresolved problem in neurobiology research and an unmet medical need. Directly reprogramming or converting astrocytes to neurons (AtN) in adult animals has been investigated as a potential strategy to facilitate brain and spinal cord recovery and advance fundamental biology. Conceptually, AtN strategies rely on forced expression or repression of lineage-specific transcription factors to make endogenous astrocytes become "induced neurons" (iNs), presumably without re-entering any pluripotent or multipotent states. The AtN-derived cells have been reported to manifest certain neuronal functions in vivo. However, this approach has raised many new questions and alternative explanations regarding the biological features of the end products (e.g., iNs versus neuron-like cells, neural functional changes, etc.), developmental biology underpinnings, and neurobiological essentials. For this paper per se, we proposed to draw an unconventional distinction between direct cell conversion and direct cell reprogramming, relative to somatic nuclear transfer, based on the experimental methods utilized to initiate the transformation process, aiming to promote a more in-depth mechanistic exploration. Moreover, we have summarized the current tactics employed for AtN induction, comparisons between the bench endeavors concerning outcome tangibility, and discussion of the issues of published AtN protocols. Lastly, the urgency to clearly define/devise the theoretical frameworks, cell biological bases, and bench specifics to experimentally validate primary data of AtN studies was highlighted.
Collapse
Affiliation(s)
- Rachel Dennison
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Esteban Usuga
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Harriet Chen
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Jacob Z. Paul
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Christian A. Arbelaez
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| | - Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
- Neurotrauma Recovery Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
32
|
Plug-and-Play Lymph Node-on-Chip: Secondary Tumor Modeling by the Combination of Cell Spheroid, Collagen Sponge and T-Cells. Int J Mol Sci 2023; 24:ijms24043183. [PMID: 36834594 PMCID: PMC9966643 DOI: 10.3390/ijms24043183] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/16/2023] [Accepted: 01/25/2023] [Indexed: 02/08/2023] Open
Abstract
Towards the improvement of the efficient study of drugs and contrast agents, the 3D microfluidic platforms are currently being actively developed for testing these substances and particles in vitro. Here, we have elaborated a microfluidic lymph node-on-chip (LNOC) as a tissue engineered model of a secondary tumor in lymph node (LN) formed due to the metastasis process. The developed chip has a collagen sponge with a 3D spheroid of 4T1 cells located inside, simulating secondary tumor in the lymphoid tissue. This collagen sponge has a morphology and porosity comparable to that of a native human LN. To demonstrate the suitability of the obtained chip for pharmacological applications, we used it to evaluate the effect of contrast agent/drug carrier size, on the penetration and accumulation of particles in 3D spheroids modeling secondary tumor. For this, the 0.3, 0.5 and 4 μm bovine serum albumin (BSA)/tannic acid (TA) capsules were mixed with lymphocytes and pumped through the developed chip. The capsule penetration was examined by scanning with fluorescence microscopy followed by quantitative image analysis. The results show that capsules with a size of 0.3 μm passed more easily to the tumor spheroid and penetrated inside. We hope that the device will represent a reliable alternative to in vivo early secondary tumor models and decrease the amount of in vivo experiments in the frame of preclinical study.
Collapse
|
33
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Condelipes PGM, Fontes PM, Godinho-Santos A, Brás EJS, Marques V, Afonso MB, Rodrigues CMP, Chu V, Gonçalves J, Conde JP. Towards personalized antibody cancer therapy: development of a microfluidic cell culture device for antibody selection. LAB ON A CHIP 2022; 22:4717-4728. [PMID: 36349999 DOI: 10.1039/d2lc00918h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Antibody therapy has been one of the most successful therapies for a wide range of diseases, including cancer. One way of expediting antibody therapy development is through phage display technology. Here, by screening thousands of randomly assembled peptide sequences, it is possible to identify potential therapeutic candidates. Conventional screening technologies do not accommodate perfusion through the system, as is the case of standard plate-based cultures. This leads to a poor translation of the experimental results obtained in vitro when moving to a more physiologically relevant setting, such as the case of preclinical animal models or clinical trials. Microfluidics is a technology that can improve screening efficacy by replicating more physiologically relevant conditions such as shear stress. In this work, a polydimethylsiloxane/polystyrene-based microfluidic system for a continuously perfused culture of cancer cells is reported. Human colorectal adenocarcinoma cells (HCT116) expressing CXCR4 were used as a cell target. Fluorescently labeled M13 phages anti-CXCR4 were used to study the efficiency of the microfluidic system as a tool to study the binding kinetics of the engineered bacteriophages. Using our microfluidic platform, we estimated a dissociation constant of 0.45 pM for the engineered phage. Additionally, a receptor internalization assay was developed using SDF-1α to verify phage specificity to the CXCR4 receptor. Upon receptor internalization there was a signal reduction, proving that the anti-CXCR4 fluorescently labelled M13 phages bound specifically to the CXCR4 receptor. The simplicity and ease of use of the microfluidic device design presented in this work can form the basis of a generic platform that facilitates the study and optimization of therapies based on interaction with biological entities such as mammalian cells.
Collapse
Affiliation(s)
- Pedro G M Condelipes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| | - Pedro Mendes Fontes
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Godinho-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eduardo J S Brás
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Marta B Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
| | - João Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores - Microsistemas e Nanotecnologias (INESC MN), Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
35
|
Huang R, Li S, Tian C, Zhou P, Zhao H, Xie W, Xiao J, Wang L, Habimana JDD, Lin Z, Yang Y, Cheng N, Li Z. Thermal stress involved in TRPV2 promotes tumorigenesis through the pathways of HSP70/27 and PI3K/Akt/mTOR in esophageal squamous cell carcinoma. Br J Cancer 2022; 127:1424-1439. [PMID: 35896815 PMCID: PMC9553907 DOI: 10.1038/s41416-022-01896-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/28/2022] [Accepted: 06/10/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The transient receptor potential vanilloid receptor 2 (TRPV2) has been found to participate in the pathogenesis of various types of cancers, however, its role(s) in the tumorigenesis of ESCC remain poorly understood. METHODS Western blotting and immunohistochemistry were performed to determine the expression profiles of TRPV2 in the ESCC patient tissues. A series of in vitro and in vivo experiments were conducted to reveal the role of TRPV2 in the tumorigenesis of ESCC. RESULTS Our study first uncovered that the activation of TRPV2 by recurrent acute thermal stress (54 °C) or O1821 (20 μM) promoted cancerous behaviours in ESCC cells. The pro-angiogenic capacity of the ESCC cells was found to be enhanced profoundly and both tumour formation and metastasis that originated from the cells were substantially promoted in nude mouse models upon the activation of TRPV2. These effects were inhibited significantly by tranilast (120 μM) and abolished by TRPV2 knockout. Conversely, overexpression of TRPV2 could switch the cells to tumorigenesis upon activation of TRPV2. Mechanistically, the driving role of TRPV2 in the progression of ESCC is mainly regulated by the HSP70/27 and PI3K/Akt/mTOR signalling pathways. CONCLUSIONS We revealed that TRPV2-PI3K/Akt/mTOR is a novel and promising target for the prevention and treatment of ESCC.
Collapse
Affiliation(s)
- Rongqi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chao Tian
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Peng Zhou
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Huifang Zhao
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Xie
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
- Department of Hepatobiliary Surgery, Provincial Cancer Hospital of Hunan, Changsha, China
| | - Jie Xiao
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Ling Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jean de Dieu Habimana
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zuoxian Lin
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yuchen Yang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Na Cheng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Zhiyuan Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
36
|
McCloskey MC, Zhang VZ, Ahmad SD, Walker S, Romanick SS, Awad HA, McGrath JL. Sourcing cells for in vitro models of human vascular barriers of inflammation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:979768. [PMID: 36483299 PMCID: PMC9724237 DOI: 10.3389/fmedt.2022.979768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 07/20/2023] Open
Abstract
The vascular system plays a critical role in the progression and resolution of inflammation. The contributions of the vascular endothelium to these processes, however, vary with tissue and disease state. Recently, tissue chip models have emerged as promising tools to understand human disease and for the development of personalized medicine approaches. Inclusion of a vascular component within these platforms is critical for properly evaluating most diseases, but many models to date use "generic" endothelial cells, which can preclude the identification of biomedically meaningful pathways and mechanisms. As the knowledge of vascular heterogeneity and immune cell trafficking throughout the body advances, tissue chip models should also advance to incorporate tissue-specific cells where possible. Here, we discuss the known heterogeneity of leukocyte trafficking in vascular beds of some commonly modeled tissues. We comment on the availability of different tissue-specific cell sources for endothelial cells and pericytes, with a focus on stem cell sources for the full realization of personalized medicine. We discuss sources available for the immune cells needed to model inflammatory processes and the findings of tissue chip models that have used the cells to studying transmigration.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samantha S. Romanick
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
37
|
Tan SY, Jing Q, Leung Z, Xu Y, Cheng LKW, Tam SST, Wu AR. Transcriptomic analysis of 3D vasculature-on-a-chip reveals paracrine factors affecting vasculature growth and maturation. LAB ON A CHIP 2022; 22:3885-3897. [PMID: 36093896 DOI: 10.1039/d2lc00570k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In vitro models of vasculature are of great importance for modelling vascular physiology and pathology. However, there is usually a lack of proper spatial patterning of interacting heterotypic cells in conventional vasculature dish models, which might confound results between contact and non-contact interactions. We use a microfluidic platform with structurally defined separation between human microvasculature and fibroblasts to probe their dynamic, paracrine interactions. We also develop a novel, versatile technique to retrieve cells embedded in extracellular matrix from the microfluidic device for downstream transcriptomic analysis, and uncover growth factor and cytokine expression profiles associated with improved vasculature growth. Paired receptor-ligand analysis further reveals paracrine signaling molecules that could be supplemented into the medium for vasculatures models where fibroblast coculture is undesirable or infeasible. These findings also provide deeper insights into the molecular cues for more physiologically relevant vascular mimicry and vascularized organoid model for clinical applications such as drug screening and disease modeling.
Collapse
Affiliation(s)
- Sin Yen Tan
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| | - Qiuyu Jing
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Ziuwin Leung
- Department of Electrical and Computer Engineering, Centre for Applied Synthetic Biology, Concordia University, Montréal, Québec H3G1M8, Canada
| | - Ying Xu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Lily Kwan Wai Cheng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Sindy Sing Ting Tam
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Angela Ruohao Wu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong S.A.R., China
| |
Collapse
|
38
|
Macaraniag C, Luan Q, Zhou J, Papautsky I. Microfluidic techniques for isolation, formation, and characterization of circulating tumor cells and clusters. APL Bioeng 2022; 6:031501. [PMID: 35856010 PMCID: PMC9288269 DOI: 10.1063/5.0093806] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cell (CTC) clusters that are shed from the primary tumor into the bloodstream are associated with a poor prognosis, elevated metastatic potential, higher proliferation rate, and distinct molecular features compared to single CTCs. Studying CTC clusters may give us information on the differences in the genetic profiles, somatic mutations, and epigenetic changes in circulating cells compared to the primary tumor and metastatic sites. Microfluidic systems offer the means of studying CTC clusters through the ability to efficiently isolate these rare cells from the whole blood of patients in a liquid biopsy. Microfluidics can also be used to develop in vitro models of CTC clusters and make possible their characterization and analysis. Ultimately, microfluidic systems can offer the means to gather insight on the complexities of the metastatic process, the biology of cancer, and the potential for developing novel or personalized therapies. In this review, we aim to discuss the advantages and challenges of the existing microfluidic systems for working with CTC clusters. We hope that an improved understanding of the role microfluidics can play in isolation, formation, and characterization of CTC clusters, which can lead to increased sophistication of microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
39
|
Guimarães CF, Cruz-Moreira D, Caballero D, Pirraco RP, Gasperini L, Kundu SC, Reis RL. Shining a Light on Cancer - Photonics in Microfluidic Tumor Modelling and Biosensing. Adv Healthc Mater 2022:e2201442. [PMID: 35998112 DOI: 10.1002/adhm.202201442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Indexed: 11/08/2022]
Abstract
Microfluidic platforms represent a powerful approach to miniaturizing important characteristics of cancers, improving in vitro testing by increasing physiological relevance. Different tools can manipulate cells and materials at the microscale, but few offer the efficiency and versatility of light and optical technologies. Moreover, light-driven technologies englobe a broad toolbox for quantifying critical biological phenomena. Herein, we review the role of photonics in microfluidic 3D cancer modeling and biosensing from three major perspectives. First, we look at optical-driven technologies that allow biomaterials and living cells to be manipulated with micro-sized precision and the opportunities to advance 3D microfluidic models by engineering cancer microenvironments' hallmarks, such as their architecture, cellular complexity, and vascularization. Second, we delve into the growing field of optofluidics, exploring how optical tools can directly interface microfluidic chips, enabling the extraction of relevant biological data, from single fluorescent signals to the complete 3D imaging of diseased cells within microchannels. Third, we review advances in optical cancer biosensing, focusing on how light-matter interactions can detect biomarkers, rare circulating tumor cells, and cell-derived structures such as exosomes. We overview photonic technologies' current challenges and caveats in microfluidic 3D cancer models, outlining future research avenues that may catapult the field. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Daniela Cruz-Moreira
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - David Caballero
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Luca Gasperini
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group -Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga and Guimarães, Portugal
| |
Collapse
|
40
|
Regmi S, Poudel C, Adhikari R, Luo KQ. Applications of Microfluidics and Organ-on-a-Chip in Cancer Research. BIOSENSORS 2022; 12:bios12070459. [PMID: 35884262 PMCID: PMC9313151 DOI: 10.3390/bios12070459] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/11/2022] [Accepted: 06/17/2022] [Indexed: 12/27/2022]
Abstract
Taking the life of nearly 10 million people annually, cancer has become one of the major causes of mortality worldwide and a hot topic for researchers to find innovative approaches to demystify the disease and drug development. Having its root lying in microelectronics, microfluidics seems to hold great potential to explore our limited knowledge in the field of oncology. It offers numerous advantages such as a low sample volume, minimal cost, parallelization, and portability and has been advanced in the field of molecular biology and chemical synthesis. The platform has been proved to be valuable in cancer research, especially for diagnostics and prognosis purposes and has been successfully employed in recent years. Organ-on-a-chip, a biomimetic microfluidic platform, simulating the complexity of a human organ, has emerged as a breakthrough in cancer research as it provides a dynamic platform to simulate tumor growth and progression in a chip. This paper aims at giving an overview of microfluidics and organ-on-a-chip technology incorporating their historical development, physics of fluid flow and application in oncology. The current applications of microfluidics and organ-on-a-chip in the field of cancer research have been copiously discussed integrating the major application areas such as the isolation of CTCs, studying the cancer cell phenotype as well as metastasis, replicating TME in organ-on-a-chip and drug development. This technology’s significance and limitations are also addressed, giving readers a comprehensive picture of the ability of the microfluidic platform to advance the field of oncology.
Collapse
Affiliation(s)
- Sagar Regmi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Department of Physics, Kathmandu University, Dhulikhel 45200, Nepal;
- Research Centre for Applied Science and Technology (RECAST), Tribhuvan University, Kathmandu 44600, Nepal;
- Nepal Academy of Science and Technology (NAST), Khumaltar, Lalitpur 44700, Nepal
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Chetan Poudel
- Department of Physics, Kathmandu University, Dhulikhel 45200, Nepal;
| | - Rameshwar Adhikari
- Research Centre for Applied Science and Technology (RECAST), Tribhuvan University, Kathmandu 44600, Nepal;
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
- Correspondence:
| |
Collapse
|
41
|
High resolution microfluidic assay and probabilistic modeling reveal cooperation between T cells in tumor killing. Nat Commun 2022; 13:3111. [PMID: 35661707 PMCID: PMC9166723 DOI: 10.1038/s41467-022-30575-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/05/2022] [Indexed: 11/28/2022] Open
Abstract
Cytotoxic T cells are important components of natural anti-tumor immunity and are harnessed in tumor immunotherapies. Immune responses to tumors and immune therapy outcomes largely vary among individuals, but very few studies examine the contribution of intrinsic behavior of the T cells to this heterogeneity. Here we show the development of a microfluidic-based in vitro method to track the outcome of antigen-specific T cell activity on many individual cancer spheroids simultaneously at high spatiotemporal resolution, which we call Multiscale Immuno-Oncology on-Chip System (MIOCS). By combining parallel measurements of T cell behaviors and tumor fates with probabilistic modeling, we establish that the first recruited T cells initiate a positive feedback loop to accelerate further recruitment to the spheroid. We also provide evidence that cooperation between T cells on the spheroid during the killing phase facilitates tumor destruction. Thus, we propose that both T cell accumulation and killing function rely on collective behaviors rather than simply reflecting the sum of individual T cell activities, and the possibility to track many replicates of immune cell-tumor interactions with the level of detail our system provides may contribute to our understanding of immune response heterogeneity. Anti-cancer cytotoxic T cell responses largely vary among individuals. Here authors show, by stochastic modeling on high throughput T cell behavior and matched tumor spheroid fate data generated by a microfluidics system, that tumor killing is dependent on T cell cooperativity, which might contribute to the heterogeneity of T cell responses.
Collapse
|
42
|
Ayuso JM, Virumbrales-Muñoz M, Lang JM, Beebe DJ. A role for microfluidic systems in precision medicine. Nat Commun 2022; 13:3086. [PMID: 35654785 PMCID: PMC9163169 DOI: 10.1038/s41467-022-30384-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
Precision oncology continues to challenge the "one-size-fits-all" dogma. Under the precision oncology banner, cancer patients are screened for molecular tumor alterations that predict treatment response, ideally leading to optimal treatments. Functional assays that directly evaluate treatment efficacy on the patient's cells offer an alternative and complementary tool to improve the accuracy of precision oncology. Unfortunately, traditional Petri dish-based assays overlook much tumor complexity, limiting their potential as predictive functional biomarkers. Here, we review past applications of microfluidic systems for precision medicine and discuss the present and potential future role of functional microfluidic assays as treatment predictors.
Collapse
Affiliation(s)
- Jose M Ayuso
- Department of Dermatology, University of Wisconsin, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Joshua M Lang
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
43
|
Qin D, Gibbons AH, Ito MM, Parimalam SS, Jiang H, Enis Karahan H, Ghalei B, Yamaguchi D, Pandian GN, Sivaniah E. Structural colour enhanced microfluidics. Nat Commun 2022; 13:2281. [PMID: 35589687 PMCID: PMC9120135 DOI: 10.1038/s41467-022-29956-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/08/2022] [Indexed: 01/11/2023] Open
Abstract
Advances in microfluidic technology towards flexibility, transparency, functionality, wearability, scale reduction or complexity enhancement are currently limited by choices in materials and assembly methods. Organized microfibrillation is a method for optically printing well-defined porosity into thin polymer films with ultrahigh resolution. Here we demonstrate this method to create self-enclosed microfluidic devices with a few simple steps, in a number of flexible and transparent formats. Structural colour, a property of organized microfibrillation, becomes an intrinsic feature of these microfluidic devices, enabling in-situ sensing capability. Since the system fluid dynamics are dependent on the internal pore size, capillary flow is shown to become characterized by structural colour, while independent of channel dimension, irrespective of whether devices are printed at the centimetre or micrometre scale. Moreover, the capability of generating and combining different internal porosities enables the OM microfluidics to be used for pore-size based applications, as demonstrated by separation of biomolecular mixtures.
Collapse
Affiliation(s)
- Detao Qin
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Andrew H Gibbons
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Masateru M Ito
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan.
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan.
| | | | - Handong Jiang
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - H Enis Karahan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Behnam Ghalei
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Daisuke Yamaguchi
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Ganesh N Pandian
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan
| | - Easan Sivaniah
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University of Advanced Study, Kyoto University, 606-8501, Kyoto, Japan.
- Department of Molecular Engineering, Kyoto University, 616-8510, Kyoto, Japan.
| |
Collapse
|
44
|
Mehta P, Rahman Z, Ten Dijke P, Boukany PE. Microfluidics meets 3D cancer cell migration. Trends Cancer 2022; 8:683-697. [PMID: 35568647 DOI: 10.1016/j.trecan.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
Abstract
An early step of metastasis requires a complex and coordinated migration of invasive tumor cells into the surrounding tumor microenvironment (TME), which contains extracellular matrix (ECM). It is being appreciated that 3D matrix-based microfluidic models have an advantage over conventional in vitro and animal models to study tumor progression events. Recent microfluidic models have enabled recapitulation of key mechanobiological features present within the TME to investigate collective cancer cell migration and invasion. Microfluidics also allows for functional interrogation and therapeutic manipulation of specific steps to study the dynamic aspects of tumor progression. In this review, we focus on recent developments in cancer cell migration and how microfluidic strategies have evolved to address the physiological complexities of the TME to visualize migration modes adapted by various tumor cells.
Collapse
Affiliation(s)
- Pranav Mehta
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
45
|
Choi JR, Kozalak G, di Bari I, Babar Q, Niknam Z, Rasmi Y, Yong KW. In Vitro Human Cancer Models for Biomedical Applications. Cancers (Basel) 2022; 14:2284. [PMID: 35565413 PMCID: PMC9099454 DOI: 10.3390/cancers14092284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/16/2022] [Accepted: 04/30/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and its incidence is steadily increasing. Although years of research have been conducted on cancer treatment, clinical treatment options for cancers are still limited. Animal cancer models have been widely used for studies of cancer therapeutics, but these models have been associated with many concerns, including inaccuracy in the representation of human cancers, high cost and ethical issues. Therefore, in vitro human cancer models are being developed quickly to fulfill the increasing demand for more relevant models in order to get a better knowledge of human cancers and to find novel treatments. This review summarizes the development of in vitro human cancer models for biomedical applications. We first review the latest development in the field by detailing various types of in vitro human cancer models, including transwell-based models, tumor spheroids, microfluidic tumor-microvascular systems and scaffold-based models. The advantages and limitations of each model, as well as their biomedical applications, are summarized, including therapeutic development, assessment of tumor cell migration, metastasis and invasion and discovery of key cancer markers. Finally, the existing challenges and future perspectives are briefly discussed.
Collapse
Affiliation(s)
- Jane Ru Choi
- Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada;
| | - Gül Kozalak
- Faculty of Engineering and Natural Sciences (FENS), Sabanci University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano-Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| | - Ighli di Bari
- Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation-Nephrology, University of Bari, 70124 Bari, Italy;
| | - Quratulain Babar
- Department of Biochemistry, Government College University, Faisalabad 38000, Pakistan;
| | - Zahra Niknam
- Proteomics Research Center, Shahid Behesti University of Medical Sciences, Tehran 1983969411, Iran;
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia 5714783734, Iran
| | - Kar Wey Yong
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
| |
Collapse
|
46
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
47
|
Vitale C, Marzagalli M, Scaglione S, Dondero A, Bottino C, Castriconi R. Tumor Microenvironment and Hydrogel-Based 3D Cancer Models for In Vitro Testing Immunotherapies. Cancers (Basel) 2022; 14:1013. [PMID: 35205760 PMCID: PMC8870468 DOI: 10.3390/cancers14041013] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
In recent years, immunotherapy has emerged as a promising novel therapeutic strategy for cancer treatment. In a relevant percentage of patients, however, clinical benefits are lower than expected, pushing researchers to deeply analyze the immune responses against tumors and find more reliable and efficient tools to predict the individual response to therapy. Novel tissue engineering strategies can be adopted to realize in vitro fully humanized matrix-based models, as a compromise between standard two-dimensional (2D) cell cultures and animal tests, which are costly and hardly usable in personalized medicine. In this review, we describe the main mechanisms allowing cancer cells to escape the immune surveillance, which may play a significant role in the failure of immunotherapies. In particular, we discuss the role of the tumor microenvironment (TME) in the establishment of a milieu that greatly favors cancer malignant progression and impact on the interactions with immune cells. Then, we present an overview of the recent in vitro engineered preclinical three-dimensional (3D) models that have been adopted to resemble the interplays between cancer and immune cells and for testing current therapies and immunotherapeutic approaches. Specifically, we focus on 3D hydrogel-based tools based on different types of polymers, discussing the suitability of each of them in reproducing the TME key features based on their intrinsic or tunable characteristics. Finally, we introduce the possibility to combine the 3D models with technological fluid dynamics platforms, reproducing the dynamic complex interactions between tumor cells and immune effectors migrated in situ via the systemic circulation, pointing out the challenges that still have to be overcome for setting more predictive preclinical assays.
Collapse
Affiliation(s)
- Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| | | | - Silvia Scaglione
- React4life SRL, 16121 Genova, Italy; (M.M.); (S.S.)
- National Research Council of Italy, Institute of Electronics, Information Engineering and Telecommunications (IEIIT), 16149 Genova, Italy
| | - Alessandra Dondero
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
- IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| |
Collapse
|
48
|
Advancing Tumor Microenvironment Research by Combining Organs-on-Chips and Biosensors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:171-203. [DOI: 10.1007/978-3-031-04039-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Xie H, Appelt JW, Jenkins RW. Going with the Flow: Modeling the Tumor Microenvironment Using Microfluidic Technology. Cancers (Basel) 2021; 13:cancers13236052. [PMID: 34885161 PMCID: PMC8656483 DOI: 10.3390/cancers13236052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The clinical success of cancer immunotherapy targeting immune checkpoints (e.g., PD-1, CTLA-4) has ushered in a new era of cancer therapeutics aimed at promoting antitumor immunity in hopes of offering durable clinical responses for patients with advanced, metastatic cancer. This success has also reinvigorated interest in developing tumor model systems that recapitulate key features of antitumor immune responses to complement existing in vivo tumor models. Patient-derived tumor models have emerged in recent years to facilitate study of tumor–immune dynamics. Microfluidic technology has enabled development of microphysiologic systems (MPSs) for the evaluation of the tumor microenvironment, which have shown early promise in studying tumor–immune dynamics. Further development of microfluidic-based “tumor-on-a-chip” MPSs to study tumor–immune interactions may overcome several key challenges currently facing tumor immunology. Abstract Recent advances in cancer immunotherapy have led a paradigm shift in the treatment of multiple malignancies with renewed focus on the host immune system and tumor–immune dynamics. However, intrinsic and acquired resistance to immunotherapy limits patient benefits and wider application. Investigations into the mechanisms of response and resistance to immunotherapy have demonstrated key tumor-intrinsic and tumor-extrinsic factors. Studying complex interactions with multiple cell types is necessary to understand the mechanisms of response and resistance to cancer therapies. The lack of model systems that faithfully recapitulate key features of the tumor microenvironment (TME) remains a challenge for cancer researchers. Here, we review recent advances in TME models focusing on the use of microfluidic technology to study and model the TME, including the application of microfluidic technologies to study tumor–immune dynamics and response to cancer therapeutics. We also discuss the limitations of current systems and suggest future directions to utilize this technology to its highest potential.
Collapse
Affiliation(s)
- Hongyan Xie
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
| | - Jackson W. Appelt
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (H.X.); (J.W.A.)
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Correspondence: ; Tel.: +617-726-9372; Fax: +844-542-5959
| |
Collapse
|
50
|
Understanding and improving cellular immunotherapies against cancer: From cell-manufacturing to tumor-immune models. Adv Drug Deliv Rev 2021; 179:114003. [PMID: 34653533 DOI: 10.1016/j.addr.2021.114003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is shaped by dynamic metabolic and immune interactions between precancerous and cancerous tumor cells and stromal cells like epithelial cells, fibroblasts, endothelial cells, and hematopoietically-derived immune cells. The metabolic states of the TME, including the hypoxic and acidic niches, influence the immunosuppressive phenotypes of the stromal and immune cells, which confers resistance to both host-mediated tumor killing and therapeutics. Numerous in vitro TME platforms for studying immunotherapies, including cell therapies, are being developed. However, we do not yet understand which immune and stromal components are most critical and how much model complexity is needed to answer specific questions. In addition, scalable sourcing and quality-control of appropriate TME cells for reproducibly manufacturing these platforms remain challenging. In this regard, lessons from the manufacturing of immunomodulatory cell therapies could provide helpful guidance. Although immune cell therapies have shown unprecedented results in hematological cancers and hold promise in solid tumors, their manufacture poses significant scale, cost, and quality control challenges. This review first provides an overview of the in vivo TME, discussing the most influential cell populations in the tumor-immune landscape. Next, we summarize current approaches for cell therapies against cancers and the relevant manufacturing platforms. We then evaluate current immune-tumor models of the TME and immunotherapies, highlighting the complexity, architecture, function, and cell sources. Finally, we present the technical and fundamental knowledge gaps in both cell manufacturing systems and immune-TME models that must be addressed to elucidate the interactions between endogenous tumor immunity and exogenous engineered immunity.
Collapse
|