1
|
Özkan A, LoGrande NT, Feitor JF, Goyal G, Ingber DE. Intestinal organ chips for disease modelling and personalized medicine. Nat Rev Gastroenterol Hepatol 2024; 21:751-773. [PMID: 39192055 DOI: 10.1038/s41575-024-00968-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/29/2024]
Abstract
Alterations in intestinal structure, mechanics and physiology underlie acute and chronic intestinal conditions, many of which are influenced by dysregulation of microbiome, peristalsis, stroma or immune responses. Studying human intestinal physiology or pathophysiology is difficult in preclinical animal models because their microbiomes and immune systems differ from those of humans. Although advances in organoid culture partially overcome this challenge, intestinal organoids still lack crucial features that are necessary to study functions central to intestinal health and disease, such as digestion or fluid flow, as well as contributions from long-term effects of living microbiome, peristalsis and immune cells. Here, we review developments in organ-on-a-chip (organ chip) microfluidic culture models of the human intestine that are lined by epithelial cells and interfaced with other tissues (such as stroma or endothelium), which can experience both fluid flow and peristalsis-like motions. Organ chips offer powerful ways to model intestinal physiology and disease states for various human populations and individual patients, and can be used to gain new insight into underlying molecular and biophysical mechanisms of disease. They can also be used as preclinical tools to discover new drugs and then validate their therapeutic efficacy and safety in the same human-relevant model.
Collapse
Affiliation(s)
- Alican Özkan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Nina Teresa LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Jessica F Feitor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
2
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2024:1-21. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
3
|
Xu H, Wen J, Yang J, Zhou S, Li Y, Xu K, Li W, Li S. Tumor-microenvironment-on-a-chip: the construction and application. Cell Commun Signal 2024; 22:515. [PMID: 39438954 PMCID: PMC11515741 DOI: 10.1186/s12964-024-01884-4] [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: 08/07/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Currently, despite the vast amounts of time and money invested in cancer treatment, cancer remains one of the primary threats to human life. The primary factor contributing to the low treatment efficacy is cancer heterogeneity. The unclear molecular mechanisms underlying tumorigenesis, coupled with the complexity of human physiology, and the inability of animal models to accurately replicate the human tumor microenvironment, pose significant hurdles in the development of novel cancer therapies. Tumor-microenvironment-on-chip (TMOC) represents a research platform that integrates three-dimensional cell culture with microfluidic systems, simulating the essential components and physiological traits of the in vivo tumor microenvironment. It offers a dynamic setting within the chip system to study tumor progression, potentially heralding a breakthrough in cancer research. In this review, we will summarize the current advancements in this platform, encompassing various types of TMOCs and their applications in different types of cancer. From our perspective, the TMOC platform necessitates enhanced integration with tissue engineering techniques and microphysiological environments before it can evolve into a more refined preclinical model for cancer research.
Collapse
Affiliation(s)
- Hanzheng Xu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Jiangtao Wen
- Linfen People's Hospital, The Seventh Clinical School of Shanxi Medical University, Shanxi, 041000, China
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Shufen Zhou
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yijie Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China.
| | - Wei Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Sen Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
4
|
Baldassarri I, Tavakol DN, Graney PL, Chramiec AG, Hibshoosh H, Vunjak-Novakovic G. An engineered model of metastatic colonization of human bone marrow reveals breast cancer cell remodeling of the hematopoietic niche. Proc Natl Acad Sci U S A 2024; 121:e2405257121. [PMID: 39374382 PMCID: PMC11494322 DOI: 10.1073/pnas.2405257121] [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: 03/19/2024] [Accepted: 08/16/2024] [Indexed: 10/09/2024] Open
Abstract
Incomplete understanding of metastatic disease mechanisms continues to hinder effective treatment of cancer. Despite remarkable advancements toward the identification of druggable targets, treatment options for patients in remission following primary tumor resection remain limited. Bioengineered human tissue models of metastatic sites capable of recreating the physiologically relevant milieu of metastatic colonization may strengthen our grasp of cancer progression and contribute to the development of effective therapeutic strategies. We report the use of an engineered tissue model of human bone marrow (eBM) to identify microenvironmental cues regulating cancer cell proliferation and to investigate how triple-negative breast cancer (TNBC) cell lines influence hematopoiesis. Notably, individual stromal components of the bone marrow niche (osteoblasts, endothelial cells, and mesenchymal stem/stromal cells) were each critical for regulating tumor cell quiescence and proliferation in the three-dimensional eBM niche. We found that hematopoietic stem and progenitor cells (HSPCs) impacted TNBC cell growth and responded to cancer cell presence with a shift of HSPCs (CD34+CD38-) to downstream myeloid lineages (CD11b+CD14+). To account for tumor heterogeneity and show proof-of-concept ability for patient-specific studies, we demonstrate that patient-derived tumor organoids survive and proliferate in the eBM, resulting in distinct shifts in myelopoiesis that are similar to those observed for aggressively metastatic cell lines. We envision that this human tissue model will facilitate studies of niche-specific metastatic progression and individualized responses to treatment.
Collapse
Affiliation(s)
- Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Pamela L. Graney
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Alan G. Chramiec
- Department of Biomedical Engineering, Columbia University, New York, NY10025
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University, New York, NY10032
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Medicine, Columbia University, New York, NY10032
- College of Dental Medicine, Columbia University, New York, NY10032
| |
Collapse
|
5
|
Lee Y, Min J, Kim S, Park W, Ko J, Jeon NL. Recapitulating the Cancer-Immunity Cycle on a Chip. Adv Healthc Mater 2024:e2401927. [PMID: 39221688 DOI: 10.1002/adhm.202401927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The cancer-immunity cycle is a fundamental framework for understanding how the immune system interacts with cancer cells, balancing T cell recognition and elimination of tumors while avoiding autoimmune reactions. Despite advancements in immunotherapy, there remains a critical need to dissect each phase of the cycle, particularly the interactions among the tumor, vasculature, and immune system within the tumor microenvironment (TME). Innovative platforms such as organ-on-a-chip, organoids, and bioprinting within microphysiological systems (MPS) are increasingly utilized to enhance the understanding of these interactions. These systems meticulously replicate crucial aspects of the TME and immune responses, providing robust platforms to study cancer progression, immune evasion, and therapeutic interventions with greater physiological relevance. This review explores the latest advancements in MPS technologies for modeling various stages of the cancer-immune cycle, critically evaluating their applications and limitations in advancing the understanding of cancer-immune dynamics and guiding the development of next-generation immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yujin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehong Min
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Solbin Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooju Park
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Qureator, Inc., San Diego, CA, 92110, USA
| |
Collapse
|
6
|
Souza IDF, Vieira JPDJ, Bonifácio ED, Avelar Freitas BAD, Torres LAG. The Microenvironment of Solid Tumors: Components and Current Challenges of Tumor-on-a-Chip Models. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39007523 DOI: 10.1089/ten.teb.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Solid tumors represent the most common type of cancer in humans and are classified into sarcomas, lymphomas, and carcinomas based on the originating cells. Among these, carcinomas, which arise from epithelial and glandular cells lining the body's tissues, are the most prevalent. Around the world, a significant increase in the incidence of solid tumors is observed during recent years. In this context, efforts to discover more effective cancer treatments have led to a deeper understanding of the tumor microenvironment (TME) and its components. Currently, the interactions between cancer cells and elements of the TME are being intensely investigated. Remarkable progress in research is noted, largely owing to the development of advanced in vitro models, such as tumor-on-a-chip models that assist in understanding and ultimately discovering new effective treatments for a specific type of cancer. The purpose of this article is to provide a review of the TME and cancer cell components, along with the advances on tumor-on-a-chip models designed to mimic tumors, offering a perspective on the current state of the art. Recent studies using this kind of microdevices that reproduce the TME have allowed a better understanding of the cancer and its treatments. Nevertheless, current applications of this technology present some limitations that must be overcome to achieve a broad application by researchers looking for a deeper knowledge of cancer and new strategies to improve current therapies.
Collapse
Affiliation(s)
- Ilva de Fátima Souza
- Institute of Science and Technology, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - João Paulo de Jesus Vieira
- Institute of Science and Technology, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
- School of Medicine, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Elton Diêgo Bonifácio
- Institute of Science and Technology, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Bethânia Alves de Avelar Freitas
- Institute of Science and Technology, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
- School of Medicine, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | - Libardo Andres Gonzalez Torres
- Institute of Science and Technology, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
- School of Medicine, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| |
Collapse
|
7
|
Lopez-Vince E, Wilhelm C, Simon-Yarza T. Vascularized tumor models for the evaluation of drug delivery systems: a paradigm shift. Drug Deliv Transl Res 2024; 14:2216-2241. [PMID: 38619704 PMCID: PMC11208221 DOI: 10.1007/s13346-024-01580-3] [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: 03/13/2024] [Indexed: 04/16/2024]
Abstract
As the conversion rate of preclinical studies for cancer treatment is low, user-friendly models that mimic the pathological microenvironment and drug intake with high throughput are scarce. Animal models are key, but an alternative to reduce their use would be valuable. Vascularized tumor-on-chip models combine great versatility with scalable throughput and are easy to use. Several strategies to integrate both tumor and vascular compartments have been developed, but few have been used to assess drug delivery. Permeability, intra/extravasation, and free drug circulation are often evaluated, but imperfectly recapitulate the processes at stake. Indeed, tumor targeting and chemoresistance bypass must be investigated to design promising cancer therapeutics. In vitro models that would help the development of drug delivery systems (DDS) are thus needed. They would allow selecting good candidates before animal studies based on rational criteria such as drug accumulation, diffusion in the tumor, and potency, as well as absence of side damage. In this review, we focus on vascularized tumor models. First, we detail their fabrication, and especially the materials, cell types, and coculture used. Then, the different strategies of vascularization are described along with their classical applications in intra/extravasation or free drug assessment. Finally, current trends in DDS for cancer are discussed with an overview of the current efforts in the domain.
Collapse
Affiliation(s)
- Elliot Lopez-Vince
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
- Université Paris Cité, Université Sorbonne Paris Nord, LVTS Inserm U1148, 75018, Paris, France
| | - Claire Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005, Paris, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, LVTS Inserm U1148, 75018, Paris, France.
| |
Collapse
|
8
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
9
|
Hsu YH, Yang WC, Chen YT, Lin CY, Yang CF, Liu WW, Shivani S, Li PC. Spatially controlled diffusion range of tumor-associated angiogenic factors to develop a tumor model using a microfluidic resistive circuit. LAB ON A CHIP 2024; 24:2644-2657. [PMID: 38576341 DOI: 10.1039/d3lc00891f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Developing a tumor model with vessels has been a challenge in microfluidics. This difficulty is because cancer cells can overgrow in a co-culture system. The up-regulation of anti-angiogenic factors during the initial tumor development can hinder neovascularization. The standard method is to develop a quiescent vessel network before loading a tumor construct in an adjacent chamber, which simulates the interaction between a tumor and its surrounding vessels. Here, we present a new method that allows a vessel network and a tumor to develop simultaneously in two linked chambers. The physiological environment of these two chambers is controlled by a microfluidic resistive circuit using two symmetric long microchannels. Applying the resistive circuit, a diffusion-dominated environment with a small 2-D pressure gradient is created across the two chambers with velocity <10.9 nm s-1 and Péclet number <6.3 × 10-5. This 2-D pressure gradient creates a V-shaped velocity clamp to confine the tumor-associated angiogenic factors at pores between the two chambers, and it has two functions. At the early stage, vasculogenesis is stimulated to grow a vessel network in the vessel chamber with minimal influence from the tumor that is still developed in the adjacent chamber. At the post-tumor-development stage, the induced steep concentration gradient at pores mimics vessel-tumor interactions to stimulate angiogenesis to grow vessels toward the tumor. Applying this method, we demonstrate that vasculogenic vessels can grow first, followed by stimulating angiogenesis. Angiogenic vessels can grow into stroma tissue up to 1.3 mm long, and vessels can also grow into or wrap around a 625 μm tumor spheroid or a tumor tissue developed from a cell suspension. In summary, our study suggests that the interactions between a developing vasculature and a growing tumor must be controlled differently throughout the tissue development process, including at the early stage when vessels are still forming and at the later stage when the tumor needs to interact with the vessels.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
- Graduate School of Advanced Technology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Wen-Chih Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Yi-Ting Chen
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Chiou-Fong Yang
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C.
| | - Wei-Wen Liu
- Graduate Institute of Oral Biology, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei 10617, Taiwan, R.O.C
| | - Subhashree Shivani
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| | - Pai-Chi Li
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, R.O.C
| |
Collapse
|
10
|
Jouybar M, de Winde CM, Wolf K, Friedl P, Mebius RE, den Toonder JMJ. Cancer-on-chip models for metastasis: importance of the tumor microenvironment. Trends Biotechnol 2024; 42:431-448. [PMID: 37914546 DOI: 10.1016/j.tibtech.2023.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
Cancer-on-chip (CoC) models, based on microfluidic chips harboring chambers for 3D tumor-cell culture, enable us to create a controlled tumor microenvironment (TME). CoC models are therefore increasingly used to systematically study effects of the TME on the various steps in cancer metastasis. Moreover, CoC models have great potential for developing novel cancer therapies and for predicting patient-specific response to cancer treatments. We review recent developments in CoC models, focusing on three main TME components: (i) the anisotropic extracellular matrix (ECM) architectures, (ii) the vasculature, and (iii) the immune system. We aim to provide guidance to biologists to choose the best CoC approach for addressing questions about the role of the TME in metastasis, and to inspire engineers to develop novel CoC technologies.
Collapse
Affiliation(s)
- Mohammad Jouybar
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Charlotte M de Winde
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands
| | - Katarina Wolf
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, Utrecht, The Netherlands
| | - Reina E Mebius
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory diseases, Amsterdam, The Netherlands
| | - Jaap M J den Toonder
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands.
| |
Collapse
|
11
|
Konopka J, Żuchowska A, Jastrzębska E. Vascularized tumor-on-chip microplatforms for the studies of neovasculature as hope for more effective cancer treatments. Biosens Bioelectron 2024; 249:115986. [PMID: 38194813 DOI: 10.1016/j.bios.2023.115986] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Abstract
Angiogenesis is the development of new blood vessels from pre-existing vasculature. Multiple factors control its course. Disorders of the distribution of angiogenic agents are responsible for development of solid tumors and its metastases. Understanding of the molecular interactions regulating pathological angiogenesis will allow for development of more effective, even personalized treatment. A simulation of angiogenesis under microflow conditions is a promising alternative to previous studies conducted on animals and on 2D cell cultures. In this review, we summarize what has been discovered so far in the field of vascularized tumor-on-a-chip platforms. For this purpose, we describe different vascularization techniques used in microfluidics, present various attempts to induce angiogenesis-on-a-chip and report some approaches to recapitulate vascularized tumor microenvironment under microflow conditions.
Collapse
Affiliation(s)
- Joanna Konopka
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Agnieszka Żuchowska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Warsaw University of Technology, Faculty of Chemistry, Medical Biotechnology, 00-664, Warsaw, Poland.
| |
Collapse
|
12
|
Esparza A, Jimenez N, Borrego EA, Browne S, Natividad-Diaz SL. Review: Human stem cell-based 3D in vitro angiogenesis models for preclinical drug screening applications. Mol Biol Rep 2024; 51:260. [PMID: 38302762 PMCID: PMC10834608 DOI: 10.1007/s11033-023-09048-2] [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: 04/10/2023] [Accepted: 11/06/2023] [Indexed: 02/03/2024]
Abstract
Vascular diseases are the underlying pathology in many life-threatening illnesses. Human cellular and molecular mechanisms involved in angiogenesis are complex and difficult to study in current 2D in vitro and in vivo animal models. Engineered 3D in vitro models that incorporate human pluripotent stem cell (hPSC) derived endothelial cells (ECs) and supportive biomaterials within a dynamic microfluidic platform provide a less expensive, more controlled, and reproducible platform to better study angiogenic processes in response to external chemical or physical stimulus. Current studies to develop 3D in vitro angiogenesis models aim to establish single-source systems by incorporating hPSC-ECs into biomimetic extracellular matrices (ECM) and microfluidic devices to create a patient-specific, physiologically relevant platform that facilitates preclinical study of endothelial cell-ECM interactions, vascular disease pathology, and drug treatment pharmacokinetics. This review provides a detailed description of the current methods used for the directed differentiation of human stem cells to endothelial cells and their use in engineered 3D in vitro angiogenesis models that have been developed within the last 10 years.
Collapse
Affiliation(s)
- Aibhlin Esparza
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Nicole Jimenez
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Edgar A Borrego
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA
| | - Shane Browne
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons, Dublin, Ireland
- CÚRAM, Centre for Research in Medical Devices, University of Galway, Galway, H91 W2TY, Ireland
| | - Sylvia L Natividad-Diaz
- Department of Metallurgical, Materials, and Biomedical Engineering (MMBME), The University of Texas at El Paso (UTEP), El Paso, TX, USA.
- 3D Printed Microphysiological Systems Laboratory, The University of Texas at El Paso, El Paso, TX, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA.
| |
Collapse
|
13
|
Hou S, Zhao Y, Chen J, Lin Y, Qi X. Tumor-associated macrophages in colorectal cancer metastasis: molecular insights and translational perspectives. J Transl Med 2024; 22:62. [PMID: 38229160 PMCID: PMC10792812 DOI: 10.1186/s12967-024-04856-x] [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: 08/11/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Metastasis is the leading cause of high mortality in colorectal cancer (CRC), which is not only driven by changes occurring within the tumor cells, but is also influenced by the dynamic interaction between cancer cells and components in the tumor microenvironment (TME). Currently, the exploration of TME remodeling and its impact on CRC metastasis has attracted increasing attention owing to its potential to uncover novel therapeutic avenues. Noteworthy, emerging studies suggested that tumor-associated macrophages (TAMs) within the TME played important roles in CRC metastasis by secreting a variety of cytokines, chemokines, growth factors and proteases. Moreover, TAMs are often associated with poor prognosis and drug resistance, making them promising targets for CRC therapy. Given the prognostic and clinical value of TAMs, this review provides an updated overview on the origin, polarization and function of TAMs, and discusses the mechanisms by which TAMs promote the metastatic cascade of CRC. Potential TAM-targeting techniques for personalized theranostics of metastatic CRC are emphasized. Finally, future perspectives and challenges for translational applications of TAMs in CRC development and metastasis are proposed to help develop novel TAM-based strategies for CRC precision medicine and holistic healthcare.
Collapse
Affiliation(s)
- Siyu Hou
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Yuanchun Zhao
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Jiajia Chen
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215011, China
| | - Yuxin Lin
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China.
| | - Xin Qi
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, 215011, China.
| |
Collapse
|
14
|
Lee S, Ki CS. Proteolytically degradable PEG hydrogel matrix mimicking tumor immune microenvironment for 3D co-culture of lung adenocarcinoma cells and macrophages. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1981-1999. [PMID: 37071065 DOI: 10.1080/09205063.2023.2204778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 04/19/2023]
Abstract
Tumor-associated macrophages and monocytes are the major stromal cell types found in the tumor immune microenvironment (TIME), which modulates tumor progression, invasion, and chemoresistance. To address the need for an in vitro three-dimensional tumor model for understanding the complex cellular interactions within the TIME, we propose a TIME-mimetic co-culture matrix composed of photo-crosslinked poly(ethylene glycol) hydrogels mimicking the characteristics of the tumor and stroma. Desmoplasia-mimetic microgels encapsulating lung adenocarcinoma cells (A549) were embedded with monocyte- or macrophage-type U937 cells in normal stroma-mimetic hydrogel, increasing the proximity between the two cell types. By modulating the proteolytic degradability of the hydrogels, we could separate different cell types with high purities for use in orthogonal assays. In addition, we demonstrated that U937 cells had distinct influences on A549 cell death depending on their activation states (i.e. monocyte, M0, or M1 phenotype). M1 macrophages suppressed tumor growth and increased the susceptibility of A549 cells to cisplatin. In contrast, monocytes upregulated cancer stem cell markers (OCT4, SOX2, and SHH) of A549 cells, showing M2-like features, such as downregulated expression of proinflammatory markers (IL6 and TNFα). These findings suggest that this co-culture system is potentially used for investigation of heterotypic cellular interactions in the TIME.
Collapse
Affiliation(s)
- Sora Lee
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, Republic of Korea
- Department of Medical Device Evaluation, National Institute of Food and Drug Safety Evaluation, Cheongju, Republic of Korea
| | - Chang Seok Ki
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, Republic of Korea
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Bouquerel C, Dubrova A, Hofer I, Phan DTT, Bernheim M, Ladaigue S, Cavaniol C, Maddalo D, Cabel L, Mechta-Grigoriou F, Wilhelm C, Zalcman G, Parrini MC, Descroix S. Bridging the gap between tumor-on-chip and clinics: a systematic review of 15 years of studies. LAB ON A CHIP 2023; 23:3906-3935. [PMID: 37592893 DOI: 10.1039/d3lc00531c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.
Collapse
Affiliation(s)
- Charlotte Bouquerel
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Anastasiia Dubrova
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Isabella Hofer
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Duc T T Phan
- Biomedicine Design, Pfizer Inc., San Diego, CA, USA
| | - Moencopi Bernheim
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Ségolène Ladaigue
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Charles Cavaniol
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Danilo Maddalo
- Department of Translational Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Luc Cabel
- Institut Curie, Department of Medical Oncology, 26 rue d'Ulm, 75005, Paris, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Claire Wilhelm
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Gérard Zalcman
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Université Paris Cité, Thoracic Oncology Department, INSERM CIC1425, Bichat Hospital, Cancer Institute AP-HP. Nord, Paris, France.
| | - Maria Carla Parrini
- Stress and Cancer Laboratory, Inserm, U830, Institut Curie, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
| | - Stéphanie Descroix
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| |
Collapse
|
16
|
Gil JF, Moura CS, Silverio V, Gonçalves G, Santos HA. Cancer Models on Chip: Paving the Way to Large-Scale Trial Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300692. [PMID: 37103886 DOI: 10.1002/adma.202300692] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/05/2023] [Indexed: 06/19/2023]
Abstract
Cancer kills millions of individuals every year all over the world (Global Cancer Observatory). The physiological and biomechanical processes underlying the tumor are still poorly understood, hindering researchers from creating new, effective therapies. Inconsistent results of preclinical research, in vivo testing, and clinical trials decrease drug approval rates. 3D tumor-on-a-chip (ToC) models integrate biomaterials, tissue engineering, fabrication of microarchitectures, and sensory and actuation systems in a single device, enabling reliable studies in fundamental oncology and pharmacology. This review includes a critical discussion about their ability to reproduce the tumor microenvironment (TME), the advantages and drawbacks of existing tumor models and architectures, major components and fabrication techniques. The focus is on current materials and micro/nanofabrication techniques used to manufacture reliable and reproducible microfluidic ToC models for large-scale trial applications.
Collapse
Affiliation(s)
- João Ferreira Gil
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Carla Sofia Moura
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, Marinha Grande, 2430-028, Portugal
- Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, 3045-093, Portugal
| | - Vania Silverio
- INESC Microsistemas e Nanotecnologias (INESC MN), Rua Alves Redol 9, Lisbon, 1000-029, Portugal
- Department of Physics, Instituto Superior Técnico, Lisbon, 1049-001, Portugal
- Associate Laboratory Institute for Health and Bioeconomy - i4HB, Lisbon, Portugal
| | - Gil Gonçalves
- TEMA, Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Aveiro, 3810-193, Portugal
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- W.J. Korf Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| |
Collapse
|
17
|
Rollins ZA, Faller R, George SC. A dynamic biomimetic model of the membrane-bound CD4-CD3-TCR complex during pMHC disengagement. Biophys J 2023; 122:3133-3145. [PMID: 37381600 PMCID: PMC10432225 DOI: 10.1016/j.bpj.2023.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
The coordinated (dis)engagement of the membrane-bound T cell receptor (TCR)-CD3-CD4 complex from the peptide-major histocompatibility complex (pMHC) is fundamental to TCR signal transduction and T cell effector function. As such, an atomic-scale understanding would not only enhance our basic understanding of the adaptive immune response but would also accelerate the rational design of TCRs for immunotherapy. In this study, we explore the impact of the CD4 coreceptor on the TCR-pMHC (dis)engagement by constructing a molecular-level biomimetic model of the CD3-TCR-pMHC and CD4-CD3-TCR-pMHC complexes within a lipid bilayer. After allowing the system complexes to equilibrate (engage), we use steered molecular dynamics to dissociate (disengage) the pMHC. We find that 1) the CD4 confines the pMHC closer to the T cell by 1.8 nm at equilibrium; 2) CD4 confinement shifts the TCR along the MHC binding groove engaging a different set of amino acids and enhancing the TCR-pMHC bond lifetime; 3) the CD4 translocates under load increasing the interaction strength between the CD4-pMHC, CD4-TCR, and CD4-CD3; and 4) upon dissociation, the CD3-TCR complex undergoes structural oscillation and increased energetic fluctuation between the CD3-TCR and CD3-lipids. These atomic-level simulations provide mechanistic insight on how the CD4 coreceptor impacts TCR-pMHC (dis)engagement. More specifically, our results provide further support (enhanced bond lifetime) for a force-dependent kinetic proofreading model and identify an alternate set of amino acids in the TCR that dominate the TCR-pMHC interaction and could thus impact the design of TCRs for immunotherapy.
Collapse
Affiliation(s)
- Zachary A Rollins
- Department of Chemical Engineering, University of California, Davis, Davis, California
| | - Roland Faller
- Department of Chemical Engineering, University of California, Davis, Davis, California
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, California.
| |
Collapse
|
18
|
Li C, Holman JB, Shi Z, Qiu B, Ding W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater Today Bio 2023; 21:100724. [PMID: 37483380 PMCID: PMC10359640 DOI: 10.1016/j.mtbio.2023.100724] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Tumor evolution is the accumulation of various tumor cell behaviors from tumorigenesis to tumor metastasis and is regulated by the tumor microenvironment (TME). However, the mechanism of solid tumor progression has not been completely elucidated, and thus, the development of tumor therapy is still limited. Recently, Tumor chips constructed by culturing tumor cells and stromal cells on microfluidic chips have demonstrated great potential in modeling solid tumors and visualizing tumor cell behaviors to exploit tumor progression. Herein, we review the methods of developing engineered solid tumors on microfluidic chips in terms of tumor types, cell resources and patterns, the extracellular matrix and the components of the TME, and summarize the recent advances of microfluidic chips in demonstrating tumor cell behaviors, including proliferation, epithelial-to-mesenchymal transition, migration, intravasation, extravasation and immune escape of tumor cells. We also outline the combination of tumor organoids and microfluidic chips to elaborate tumor organoid-on-a-chip platforms, as well as the practical limitations that must be overcome.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Joseph Benjamin Holman
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
19
|
Yau JNN, Adriani G. Three-dimensional heterotypic colorectal cancer spheroid models for evaluation of drug response. Front Oncol 2023; 13:1148930. [PMID: 37469395 PMCID: PMC10352797 DOI: 10.3389/fonc.2023.1148930] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/15/2023] [Indexed: 07/21/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide. Improved preclinical tumor models are needed to make treatment screening clinically relevant and address disease mortality. Advancements in 3D cell culture have enabled a greater recapitulation of the architecture and heterogeneity of the tumor microenvironment (TME). This has enhanced their pathophysiological relevance and enabled more accurate predictions of tumor progression and drug response in patients. An increasing number of 3D CRC spheroid models include cell populations such as cancer-associated fibroblasts (CAFs), endothelial cells (ECs), immune cells, and gut bacteria to better mimic the in vivo regulation of signaling pathways. Furthermore, cell heterogeneity within the 3D spheroid models enables the identification of new therapeutic targets to develop alternative treatments and test TME-target therapies. In this mini review, we present the advances in mimicking tumor heterogeneity in 3D CRC spheroid models by incorporating CAFs, ECs, immune cells, and gut bacteria. We introduce how, in these models, the diverse cells influence chemoresistance and tumor progression of the CRC spheroids. We also highlight important parameters evaluated during drug screening in the CRC heterocellular spheroids.
Collapse
Affiliation(s)
- Jia Ning Nicolette Yau
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
20
|
Shirure VS, Yechikov S, Shergill BS, Dehghani T, Block AV, Sodhi H, Panitch A, George SC. Mitigating neutrophil trafficking and cardiotoxicity with DS-IkL in a microphysiological system of a cytokine storm. LAB ON A CHIP 2023; 23:3050-3061. [PMID: 37278194 PMCID: PMC10330849 DOI: 10.1039/d2lc01070d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A feature of severe COVID-19 is the onset of an acute and intense systemic inflammatory response referred to as the "cytokine storm". The cytokine storm is characterized by high serum levels of inflammatory cytokines and the subsequent transport of inflammatory cells to damaging levels in vital organs (e.g., myocarditis). Immune trafficking and its effect on underlying tissues (e.g., myocardium) are challenging to observe at a high spatial and temporal resolution in mouse models. In this study, we created a vascularized organ-on-a-chip system to mimic cytokine storm-like conditions and tested the effectiveness of a novel multivalent selectin-targeting carbohydrate conjugate (composed of DS - dermatan sulfate and IkL - a selectin-binding peptide, termed DS-IkL) in blocking infiltration of polymorphonuclear leukocytes (PMN). Our data shows that cytokine storm-like conditions induce endothelial cells to produce additional inflammatory cytokines and facilitate infiltration of PMNs into tissue. Treatment of tissues with DS-IkL (60 μM) reduced PMN accumulation in the tissue by >50%. We then created cytokine storm-like conditions in a vascularized cardiac tissue-chip and found that PMN infiltration increases the spontaneous beating rate of the cardiac tissue, and this effect is eliminated by treatment with DS-IkL (60 μM). In summary, we demonstrate the utility of an organ-on-a-chip platform to mimic COVID-19 related cytokine storm and that blocking leukocyte infiltration with DS-IkL could be a viable strategy to mitigate associated cardiac complications.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Sergey Yechikov
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Bhupinder S Shergill
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Tima Dehghani
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Anton V Block
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Harkanwalpreet Sodhi
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA 95616, USA.
| |
Collapse
|
21
|
Wan Z, Floryan MA, Coughlin MF, Zhang S, Zhong AX, Shelton SE, Wang X, Xu C, Barbie DA, Kamm RD. New Strategy for Promoting Vascularization in Tumor Spheroids in a Microfluidic Assay. Adv Healthc Mater 2023; 12:e2201784. [PMID: 36333913 PMCID: PMC10156888 DOI: 10.1002/adhm.202201784] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Previous studies have developed vascularized tumor spheroid models to demonstrate the impact of intravascular flow on tumor progression and treatment. However, these models have not been widely adopted so the vascularization of tumor spheroids in vitro is generally lower than vascularized tumor tissues in vivo. To improve the tumor vascularization level, a new strategy is introduced to form tumor spheroids by adding fibroblasts (FBs) sequentially to a pre-formed tumor spheroid and demonstrate this method with tumor cell lines from kidney, lung, and ovary cancer. Tumor spheroids made with the new strategy have higher FB densities on the periphery of the tumor spheroid, which tend to enhance vascularization. The vessels close to the tumor spheroid made with this new strategy are more perfusable than the ones made with other methods. Finally, chimeric antigen receptor (CAR) T cells are perfused under continuous flow into vascularized tumor spheroids to demonstrate immunotherapy evaluation using vascularized tumor-on-a-chip model. This new strategy for establishing tumor spheroids leads to increased vascularization in vitro, allowing for the examination of immune, endothelial, stromal, and tumor cell responses under static or flow conditions.
Collapse
Affiliation(s)
- Zhengpeng Wan
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMA02215USA
| | - Marie A. Floryan
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark F. Coughlin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Shun Zhang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Amy X. Zhong
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah E. Shelton
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMA02215USA
| | - Xun Wang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Chenguang Xu
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
| | - David A. Barbie
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMA02215USA
| | - Roger D. Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
22
|
Phuangbubpha P, Thara S, Sriboonaied P, Saetan P, Tumnoi W, Charoenpanich A. Optimizing THP-1 Macrophage Culture for an Immune-Responsive Human Intestinal Model. Cells 2023; 12:1427. [PMID: 37408263 DOI: 10.3390/cells12101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
Previously established immune-responsive co-culture models with macrophages have limitations due to the dedifferentiation of macrophages in long-term cultures. This study is the first report of a long-term (21-day) triple co-culture of THP-1 macrophages (THP-1m) with Caco-2 intestinal epithelial cells and HT-29-methotrexate (MTX) goblet cells. We demonstrated that high-density seeded THP-1 cells treated with 100 ng/mL phorbol 12-myristate 13-acetate for 48 h differentiated stably and could be cultured for up to 21 days. THP-1m were identified by their adherent morphology and lysosome expansion. In the triple co-culture immune-responsive model, cytokine secretions during lipopolysaccharide-induced inflammation were confirmed. Tumor necrosis factor-alpha and interleukin 6 levels were elevated in the inflamed state, reaching 824.7 ± 130.0 pg/mL and 609.7 ± 139.5 pg/mL, respectively. Intestinal membrane integrity was maintained with a transepithelial electrical resistance value of 336.4 ± 18.0 Ω·cm2. Overall, our findings suggest that THP-1m can be effectively employed in models of long-term immune responses in both normal and chronic inflammatory states of the intestinal epithelium, making them a valuable tool for future research on the association between the immune system and gut health.
Collapse
Affiliation(s)
- Pornwipa Phuangbubpha
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Sanya Thara
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Patsawee Sriboonaied
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Puretat Saetan
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Wanwiwa Tumnoi
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Adisri Charoenpanich
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
23
|
Advancing preclinical cancer models to assess clinically relevant outcomes. BMC Cancer 2023; 23:230. [PMID: 36899363 PMCID: PMC10007806 DOI: 10.1186/s12885-023-10715-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Cancer models are indispensable research tools for elucidating the mechanisms involved in tumor onset, progression and treatment resistance. They are key in evaluating therapeutics prior clinical trials. In this editorial, we invite contributions for a BMC Cancer's Collection of articles addressing 'Advances in pre-clinical cancer models' towards relivable outcomes at the preclinical stage.
Collapse
|
24
|
Huang CBX, Tu TY. Recent advances in vascularized tumor-on-a-chip. Front Oncol 2023; 13:1150332. [PMID: 37064144 PMCID: PMC10099572 DOI: 10.3389/fonc.2023.1150332] [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: 01/24/2023] [Accepted: 03/13/2023] [Indexed: 04/18/2023] Open
Abstract
The vasculature plays a critical role in cancer progression and metastasis, representing a pivotal aspect in the creation of cancer models. In recent years, the emergence of organ-on-a-chip technology has proven to be a robust tool, capable of replicating in vivo conditions with exceptional spatiotemporal resolution, making it a significant asset in cancer research. This review delves into the latest developments in 3D microfluidic vascularized tumor models and their applications in vitro, focusing on heterotypic cellular interactions, the mechanisms of metastasis, and therapeutic screening. Additionally, the review examines the benefits and drawbacks of these models, as well as the future prospects for their advancement.
Collapse
Affiliation(s)
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
- *Correspondence: Ting-Yuan Tu,
| |
Collapse
|
25
|
Rollins ZA, Chan A, Shirure VS, George SC. Receptor-ligand non-equilibrium kinetics (RLNEK) 1.0: An integrated Trackmate laminar flow chamber analysis. J Immunol Methods 2022; 511:113381. [PMID: 36341963 DOI: 10.1016/j.jim.2022.113381] [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: 02/09/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Although parallel plate flow chamber assays are widely performed, extraction of kinetic parameters is limited to specialized labs with mathematical expertise and customized video-microscopy tracking tools. The recent development of Trackmate has increased researcher accessibility to tracking particles in video-microscopy experiments; however, there is a lack of tools that analyze this tracking information. We report a software tool, compatible with Trackmate, that extracts Receptor Ligand Non-Equilibrium Kinetic (RLNEK) parameters from video-microscopy data. This software should be of particular interest to the community of researchers and scientists interrogating the target-specific binding and release of immune cells.
Collapse
Affiliation(s)
- Zachary A Rollins
- Department of Chemical Engineering, University of California, Davis, Davis, CA, USA
| | - Allison Chan
- Department of Chemical Engineering, University of California, Davis, Davis, CA, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
26
|
Curtis MB, Kelly N, Hughes CCW, George SC. Organotypic stromal cells impact endothelial cell transcriptome in 3D microvessel networks. Sci Rep 2022; 12:20434. [PMID: 36443378 PMCID: PMC9705391 DOI: 10.1038/s41598-022-24013-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Endothelial cells line all major blood vessels and serve as integral regulators of many functions including vessel diameter, cellular trafficking, and transport of soluble mediators. Despite similar functions, the phenotype of endothelial cells is highly organ-specific, yet our understanding of the mechanisms leading to organ-level differentiation is incomplete. We generated 3D microvessel networks by combining a common naïve endothelial cell with six different stromal cells derived from the lung, skin, heart, bone marrow, pancreas, and pancreatic cancer. Single cell RNA-Seq analysis of the microvessel networks reveals five distinct endothelial cell populations, for which the relative proportion depends on the stromal cell population. Morphologic features of the organotypic vessel networks inversely correlate with a cluster of endothelial cells associated with protein synthesis. The organotypic stromal cells were each characterized by a unique subpopulation of cells dedicated to extracellular matrix organization and assembly. Finally, compared to cells in 2D monolayer, the endothelial cell transcriptome from the 3D in vitro heart, skin, lung, and pancreas microvessel networks are more similar to the in vivo endothelial cells from the respective organs. We conclude that stromal cells contribute to endothelial cell and microvessel network organ tropism, and create an endothelial cell phenotype that more closely resembles that present in vivo.
Collapse
Affiliation(s)
- Matthew B Curtis
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA
| | - Natalie Kelly
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA
| | - Christopher C W Hughes
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E. Health Sciences Drive, Room 2315, Davis, CA, 95616, USA.
| |
Collapse
|
27
|
Wang R, Zhang C, Li D, Yao Y. Tumor-on-a-chip: Perfusable vascular incorporation brings new approach to tumor metastasis research and drug development. Front Bioeng Biotechnol 2022; 10:1057913. [PMID: 36483772 PMCID: PMC9722735 DOI: 10.3389/fbioe.2022.1057913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/11/2022] [Indexed: 07/21/2023] Open
Abstract
The extracellular matrix interacts with cancer cells and is a key factor in the development of cancer. Traditional two-dimensional models cannot mimic the natural in situ environment of cancer tissues, whereas three-dimensional (3D) models such as spherical culture, bioprinting, and microfluidic approaches can achieve in vitro reproduction of certain structures and components of the tumor microenvironment, including simulation of the hypoxic environment of tumor tissue. However, the lack of a perfusable vascular network is a limitation of most 3D models. Solid tumor growth and metastasis require angiogenesis, and tumor models with microvascular networks have been developed to better understand underlying mechanisms. Tumor-on-a-chip technology combines the advantages of microfluidics and 3D cell culture technology for the simulation of tumor tissue complexity and characteristics. In this review, we summarize progress in constructing tumor-on-a-chip models with efficiently perfused vascular networks. We also discuss the applications of tumor-on-a-chip technology to studying the tumor microenvironment and drug development. Finally, we describe the creation of several common tumor models based on this technology to provide a deeper understanding and new insights into the design of vascularized cancer models. We believe that the tumor-on-a-chip approach is an important development that will provide further contributions to the field.
Collapse
Affiliation(s)
| | | | - Danxue Li
- *Correspondence: Danxue Li, ; Yang Yao,
| | - Yang Yao
- *Correspondence: Danxue Li, ; Yang Yao,
| |
Collapse
|
28
|
Krysko DV, Demuynck R, Efimova I, Naessens F, Krysko O, Catanzaro E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022; 11:3705. [PMID: 36429133 PMCID: PMC9688238 DOI: 10.3390/cells11223705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic cell death (ICD) is a functionally unique form of cell death that promotes a T-cell-dependent anti-tumor immune response specific to antigens originating from dying cancer cells. Many anticancer agents and strategies induce ICD, but despite their robust effects in vitro and in vivo on mice, translation into the clinic remains challenging. A major hindrance in antitumor research is the poor predictive ability of classic 2D in vitro models, which do not consider tumor biological complexity, such as the contribution of the tumor microenvironment (TME), which plays a crucial role in immunosuppression and cancer evasion. In this review, we describe different tumor models, from 2D cultures to organ-on-a-chip technology, as well as spheroids and perfusion bioreactors, all of which mimic the different degrees of the TME complexity. Next, we discuss how 3D cell cultures can be applied to study ICD and how to increase the translational potential of the ICD inducers. Finally, novel research directions are provided regarding ICD in the 3D cellular context which may lead to novel immunotherapies for cancer.
Collapse
Affiliation(s)
- Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Robin Demuynck
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Catanzaro
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
29
|
Geyer M, Queiroz K. Microfluidic Platforms for High-Throughput Pancreatic Ductal Adenocarcinoma Organoid Culture and Drug Screening. Front Cell Dev Biol 2022; 9:761807. [PMID: 35004672 PMCID: PMC8733732 DOI: 10.3389/fcell.2021.761807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC), the most common pancreatic cancer type, is believed to become the second leading cause of cancer-related deaths by 2030 with mortality rates of up to 93%. It is often detected at a late stage due to lacking symptoms, and therefore surgical removal of the tumor is the only treatment option for patients. Only 20% of the tumors are resectable, mainly due to early metastasis. Therefore, for 80% of cases chemotherapeutic treatment is the leading therapy for patients. PDAC is characterized by high-density stroma which induces hypoxic conditions and high interstitial pressure. These factors impact carcinogenesis and progression of PDAC and support the formation of an immunosuppressive microenvironment that renders this tumor type refractory to immunotherapies. Most in vitro PDAC models have limited translational relevance, as these fail to recapitulate relevant aspects of PDAC complexity. Altogether, there is an urgent need for novel and innovative PDAC modeling platforms. Here, we discuss the relevance of microfluidic and organoid technologies as platforms for modeling bio- and physicochemical features of PDAC and as translational models that enable high-throughput phenotypic drug screenings, while also allowing for the development of novel personalized models used to identify treatment responsive patient subsets.
Collapse
|
30
|
Jones TH, Song JW, Abushahin L. Tumor treating fields: An emerging treatment modality for thoracic and abdominal cavity cancers. Transl Oncol 2022; 15:101296. [PMID: 34847422 PMCID: PMC8633677 DOI: 10.1016/j.tranon.2021.101296] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Tumor treating fields (TTFields)-an intermediate-frequency, electric field therapy-has emerged as a promising alternative therapy for the treatment of solid cancers. Since the first publication describing the anticancer effects of TTFields in 2004 there have been numerous follow-up studies by other groups, either to confirm the efficacy of TTFields or to study the primary mechanism of interaction. The overwhelming conclusion from these in vitro studies is that TTFields reduce the viability of aggressively replicating cell lines. However, there is still speculation as to the primary mechanism for this effect; moreover, observations both in vitro and in vivo of inhibited migration and metastases have been made, which may be unrelated to the originally proposed hypothesis of replication stress. Adding to this, the in vivo environment is much more complex spatially, structurally, and involves intricate networks of cell signaling, all of which could change the efficacy of TTFields in the same way pharmaceutical interventions often struggle transitioning in vivo. Despite this, TTFields have shown promise in clinical practice on multiple cancer types, which begs the question: has the primary mechanism carried over from in vitro to in vivo or are there new mechanisms at play? The goal of this review is to highlight the current proposed mechanism of action of TTFields based primarily on in vitro experiments and animal models, provide a summary of the clinical efficacy of TTFields, and finally, propose future directions of research to identify all possible mechanisms in vivo utilizing novel tumor-on-a-chip platforms.
Collapse
Affiliation(s)
- Travis H Jones
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201W. 19th Avenue, E406 Scott Laboratory, Columbus, OH 43210, United States; Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, 1800 Canon Drive, 1300G, Columbus, OH 43210, United States
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, 201W. 19th Avenue, E406 Scott Laboratory, Columbus, OH 43210, United States; Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, United States.
| | - Laith Abushahin
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, 1800 Canon Drive, 1300G, Columbus, OH 43210, United States; Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, United States.
| |
Collapse
|
31
|
Glaser DE, Curtis MB, Sariano PA, Rollins ZA, Shergill BS, Anand A, Deely AM, Shirure VS, Anderson L, Lowen JM, Ng NR, Weilbaecher K, Link DC, George SC. Organ-on-a-chip model of vascularized human bone marrow niches. Biomaterials 2022; 280:121245. [PMID: 34810038 PMCID: PMC10658812 DOI: 10.1016/j.biomaterials.2021.121245] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Bone marrow niches (endosteal and perivascular) play important roles in both normal bone marrow function and pathological processes such as cancer cell dormancy. Unraveling the mechanisms underlying these events in humans has been severely limited by models that cannot dissect dynamic events at the niche level. Utilizing microfluidic and stem cell technologies, we present a 3D in vitro model of human bone marrow that contains both the perivascular and endosteal niches, complete with dynamic, perfusable vascular networks. We demonstrate that our model can replicate in vivo bone marrow function, including maintenance and differentiation of CD34+ hematopoietic stem/progenitor cells, egress of neutrophils (CD66b+), and niche-specific responses to doxorubicin and granulocyte-colony stimulating factor. Our platform provides opportunities to accelerate current understanding of human bone marrow function and drug response with high spatial and temporal resolution.
Collapse
Affiliation(s)
- Drew E Glaser
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Matthew B Curtis
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Peter A Sariano
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Zachary A Rollins
- Department of Chemical Engineering, University of California, Davis, 1 Shields Ave, Bainer 3106, Davis, CA 95616, USA
| | - Bhupinder S Shergill
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Aravind Anand
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Alyssa M Deely
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Leif Anderson
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Jeremy M Lowen
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA
| | - Natalie R Ng
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Dr, Campus Box 1100, St Louis, MO 63130, USA
| | - Katherine Weilbaecher
- Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Campus Box 8066, St. Louis, MO 63110, USA
| | - Daniel C Link
- Department of Medicine, Washington University in St. Louis, 660 S Euclid Ave, Campus Box 8066, St. Louis, MO 63110, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, 451 E Health Sciences Dr, GBSF 2303, Davis, CA 95616, USA.
| |
Collapse
|
32
|
DePalma TJ, Sivakumar H, Skardal A. Strategies for developing complex multi-component in vitro tumor models: Highlights in glioblastoma. Adv Drug Deliv Rev 2022; 180:114067. [PMID: 34822927 PMCID: PMC10560581 DOI: 10.1016/j.addr.2021.114067] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
In recent years, many research groups have begun to utilize bioengineered in vitro models of cancer to study mechanisms of disease progression, test drug candidates, and develop platforms to advance personalized drug treatment options. Due to advances in cell and tissue engineering over the last few decades, there are now a myriad of tools that can be used to create such in vitro systems. In this review, we describe the considerations one must take when developing model systems that accurately mimic the in vivo tumor microenvironment (TME) and can be used to answer specific scientific questions. We will summarize the importance of cell sourcing in models with one or multiple cell types and outline the importance of choosing biomaterials that accurately mimic the native extracellular matrix (ECM) of the tumor or tissue that is being modeled. We then provide examples of how these two components can be used in concert in a variety of model form factors and conclude by discussing how biofabrication techniques such as bioprinting and organ-on-a-chip fabrication can be used to create highly reproducible complex in vitro models. Since this topic has a broad range of applications, we use the final section of the review to dive deeper into one type of cancer, glioblastoma, to illustrate how these components come together to further our knowledge of cancer biology and move us closer to developing novel drugs and systems that improve patient outcomes.
Collapse
Affiliation(s)
- Thomas J DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Hemamylammal Sivakumar
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
33
|
Tan ML, Ling L, Fischbach C. Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro. Adv Drug Deliv Rev 2021; 176:113852. [PMID: 34197895 PMCID: PMC8440401 DOI: 10.1016/j.addr.2021.113852] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Despite decades of research and advancements in diagnostic and treatment modalities, cancer remains a major global healthcare challenge. This is due in part to a lack of model systems that allow investigating the mechanisms underlying tumor development, progression, and therapy resistance under relevant conditions in vitro. Tumor cell interactions with their surroundings influence all stages of tumorigenesis and are shaped by both biological and biophysical cues including cell-cell and cell-extracellular matrix (ECM) interactions, tissue architecture and mechanics, and mass transport. Engineered tumor models provide promising platforms to elucidate the individual and combined contributions of these cues to tumor malignancy under controlled and physiologically relevant conditions. This review will summarize current knowledge of the biological and biophysical microenvironmental cues that influence tumor development and progression, present examples of in vitro model systems that are presently used to study these interactions and highlight advancements in tumor engineering approaches to further improve these technologies.
Collapse
Affiliation(s)
- Matthew L Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lu Ling
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
34
|
Chakraborty S, DePalma TJ, Skardal A. Increasing Accuracy of In Vitro Cancer Models: Engineering Stromal Complexity into Tumor Organoid Platforms. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Srija Chakraborty
- Department of Biomedical Engineering The Ohio State University 3022 Fontana Labs 140 W. 19th Avenue Columbus OH 43210 USA
| | - Thomas J. DePalma
- Department of Biomedical Engineering The Ohio State University 3022 Fontana Labs 140 W. 19th Avenue Columbus OH 43210 USA
| | - Aleksander Skardal
- Department of Biomedical Engineering The Ohio State University 3022 Fontana Labs 140 W. 19th Avenue Columbus OH 43210 USA
- Center for Cancer Engineering The Ohio State University and Arthur G. James Comprehensive Cancer Center Columbus OH 43210 USA
| |
Collapse
|
35
|
Del Piccolo N, Shirure VS, Bi Y, Goedegebuure SP, Gholami S, Hughes CC, Fields RC, George SC. Tumor-on-chip modeling of organ-specific cancer and metastasis. Adv Drug Deliv Rev 2021; 175:113798. [PMID: 34015419 DOI: 10.1016/j.addr.2021.05.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023]
Abstract
Every year, cancer claims millions of lives around the globe. Unfortunately, model systems that accurately mimic human oncology - a requirement for the development of more effective therapies for these patients - remain elusive. Tumor development is an organ-specific process that involves modification of existing tissue features, recruitment of other cell types, and eventual metastasis to distant organs. Recently, tissue engineered microfluidic devices have emerged as a powerful in vitro tool to model human physiology and pathology with organ-specificity. These organ-on-chip platforms consist of cells cultured in 3D hydrogels and offer precise control over geometry, biological components, and physiochemical properties. Here, we review progress towards organ-specific microfluidic models of the primary and metastatic tumor microenvironments. Despite the field's infancy, these tumor-on-chip models have enabled discoveries about cancer immunobiology and response to therapy. Future work should focus on the development of autologous or multi-organ systems and inclusion of the immune system.
Collapse
|
36
|
3D tumor spheroid microarray for high-throughput, high-content natural killer cell-mediated cytotoxicity. Commun Biol 2021; 4:893. [PMID: 34290356 PMCID: PMC8295284 DOI: 10.1038/s42003-021-02417-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has emerged as a promising approach to treating several forms of cancer. Use of immune cells, such as natural killer (NK) cells, along with small molecule drugs and antibodies through antibody dependent cell-mediated cytotoxicity (ADCC) has been investigated as a potential combination therapy for some difficult to treat solid tumors. Nevertheless, there remains a need to develop tools that support co-culture of target cancer cells and effector immune cells in a contextually relevant three-dimensional (3D) environment to provide a rapid means to screen for and optimize ADCC-drug combinations. To that end, here we have developed a high throughput 330 micropillar-microwell sandwich platform that enables 3D co-culture of NK92-CD16 cells with pancreatic (MiaPaCa-2) and breast cancer cell lines (MCF-7 and MDA-MB-231). The platform successfully mimicked hypoxic conditions found in a tumor microenvironment and was used to demonstrate NK-cell mediated cell cytotoxicity in combination with two monoclonal antibodies; Trastuzumab and Atezolizumab. The platform was also used to show dose response behavior of target cancer cells with reduced EC50 values for paclitaxel (an anti-cancer chemotherapeutic) when treated with both NK cells and antibody. Such a platform may be used to develop more personalized cancer therapies using patient-derived cancer cells.
Collapse
|
37
|
Kwee BJ, Sung KE. Engineering microenvironments for manufacturing therapeutic cells. Exp Biol Med (Maywood) 2021; 246:1845-1856. [PMID: 34250847 DOI: 10.1177/15353702211026922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There are a growing number of globally approved products and clinical trials utilizing autologous and allogeneic therapeutic cells for applications in regenerative medicine and immunotherapies. However, there is a need to develop rapid and cost-effective methods for manufacturing therapeutically effective cells. Furthermore, the resulting manufactured cells may exhibit heterogeneities that result in mixed therapeutic outcomes. Engineering approaches that can provide distinct microenvironmental cues to these cells may be able to enhance the growth and characterization of these cell products. This mini-review describes strategies to potentially enhance the expansion of therapeutic cells with biomaterials and bioreactors, as well as to characterize the cell products with microphysiological systems. These systems can provide distinct cues to maintain the quality attributes of the cells and evaluate their function in physiologically relevant conditions.
Collapse
Affiliation(s)
- Brian J Kwee
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Kyung E Sung
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| |
Collapse
|
38
|
Larionova I, Kazakova E, Gerashchenko T, Kzhyshkowska J. New Angiogenic Regulators Produced by TAMs: Perspective for Targeting Tumor Angiogenesis. Cancers (Basel) 2021; 13:cancers13133253. [PMID: 34209679 PMCID: PMC8268686 DOI: 10.3390/cancers13133253] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Since the targeting of a single pro-angiogenic factor fails to improve oncological disease outcome, significant efforts have been made to identify new pro-angiogenic factors that could compensate for the deficiency of current therapy or act independently as single drugs. Our review aims to present the state-of-the art for well-known and recently described factors produced by macrophages that induce and regulate angiogenesis. A number of positive and negative regulators of angiogenesis in the tumor microenvironment are produced by tumor-associated macrophages (TAMs). Accumulating evidence has indicated that, apart from the well-known angiogenic factors, there are plenty of novel angiogenesis-regulating proteins that belong to different classes. We summarize the data regarding the direct or indirect mechanisms of the interaction of these factors with endothelial cells during angiogenesis. We highlight the recent findings that explain the limitations in the efficiency of current anti-angiogenic therapy approaches. Abstract Angiogenesis is crucial to the supply of a growing tumor with nutrition and oxygen. Inhibition of angiogenesis is one of the main treatment strategies for colorectal, lung, breast, renal, and other solid cancers. However, currently applied drugs that target VEGF or receptor tyrosine kinases have limited efficiency, which raises a question concerning the mechanism of patient resistance to the already developed drugs. Tumor-associated macrophages (TAMs) were identified in the animal tumor models as a key inducer of the angiogenic switch. TAMs represent a potent source not only for VEGF, but also for a number of other pro-angiogenic factors. Our review provides information about the activity of secreted regulators of angiogenesis produced by TAMs. They include members of SEMA and S100A families, chitinase-like proteins, osteopontin, and SPARC. The COX-2, Tie2, and other factors that control the pro-angiogenic activity of TAMs are also discussed. We highlight how these recent findings explain the limitations in the efficiency of current anti-angiogenic therapy. Additionally, we describe genetic and posttranscriptional mechanisms that control the expression of factors regulating angiogenesis. Finally, we present prospects for the complex targeting of the pro-angiogenic activity of TAMs.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
- Correspondence: (I.L.); (J.K.)
| | - Elena Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 68167 Mannheim, Germany
- Correspondence: (I.L.); (J.K.)
| |
Collapse
|
39
|
Firatligil-Yildirir B, Bati-Ayaz G, Tahmaz I, Bilgen M, Pesen-Okvur D, Yalcin-Ozuysal O. On-chip determination of tissue-specific metastatic potential of breast cancer cells. Biotechnol Bioeng 2021; 118:3799-3810. [PMID: 34110014 DOI: 10.1002/bit.27855] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/23/2021] [Accepted: 06/07/2021] [Indexed: 11/07/2022]
Abstract
Metastasis is one of the major obstacles for breast cancer patients. Limitations of current models demand the development of custom platforms to predict metastatic potential and homing choices of cancer cells. Here, two organ-on-chip platforms, invasion/chemotaxis (IC-chip) and extravasation (EX-chip) were used for the quantitative assessment of invasion and extravasation towards specific tissues. Lung, liver and breast microenvironments were simulated in the chips using tissue-specific cells embedded in matrigel. In the IC-chip, invasive MDA-MB-231, but not noninvasive MCF-7 breast cancer cells invaded into lung and liver microenvironments. In the EX-chip, MDA-MB-231 cells extravasated more into the lung compared to the liver and breast microenvironments. In addition, lung-specific MDA-MB-231 clone invaded and extravasated into the lung microenvironment more efficiently than the bone-specific clone. Both invasion/chemotaxis and extravasation results were in agreement with published clinical data. Collectively, our results show that IC-chip and EX-chip, simulating tissue-specific microenvironments, can distinguish different in vivo metastatic phenotypes, in vitro. Determination of tissue-specific metastatic potential of breast cancer cells is expected to improve diagnosis and help select the ideal therapy.
Collapse
Affiliation(s)
| | - Gizem Bati-Ayaz
- Izmir Institute of Technology, Biotechnology and Bioengineering Graduate Program, Izmir, Turkey
| | - Ismail Tahmaz
- Izmir Institute of Technology, Biotechnology and Bioengineering Graduate Program, Izmir, Turkey
| | - Muge Bilgen
- Izmir Institute of Technology, Biotechnology and Bioengineering Graduate Program, Izmir, Turkey
| | - Devrim Pesen-Okvur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Ozden Yalcin-Ozuysal
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| |
Collapse
|
40
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
41
|
Delle Cave D, Rizzo R, Sainz B, Gigli G, del Mercato LL, Lonardo E. The Revolutionary Roads to Study Cell-Cell Interactions in 3D In Vitro Pancreatic Cancer Models. Cancers (Basel) 2021; 13:930. [PMID: 33672435 PMCID: PMC7926501 DOI: 10.3390/cancers13040930] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/07/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer, the fourth most common cancer worldwide, shows a highly unsuccessful therapeutic response. In the last 10 years, neither important advancements nor new therapeutic strategies have significantly impacted patient survival, highlighting the need to pursue new avenues for drug development discovery and design. Advanced cellular models, resembling as much as possible the original in vivo tumor environment, may be more successful in predicting the efficacy of future anti-cancer candidates in clinical trials. In this review, we discuss novel bioengineered platforms for anticancer drug discovery in pancreatic cancer, from traditional two-dimensional models to innovative three-dimensional ones.
Collapse
Affiliation(s)
- Donatella Delle Cave
- Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), Via Pietro Castellino 111, 80131 Naples, Italy;
| | - Riccardo Rizzo
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy; (R.R.); (G.G.); (L.L.d.M.)
| | - Bruno Sainz
- Department of Cancer Biology, Instituto de Investigaciones Biomedicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain;
- Spain and Chronic Diseases and Cancer, Area 3-Instituto Ramon y Cajal de Investigacion Sanitaria (IRYCIS), 28029 Madrid, Spain
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy; (R.R.); (G.G.); (L.L.d.M.)
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, via Arnesano, 73100 Lecce, Italy
| | - Loretta L. del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy; (R.R.); (G.G.); (L.L.d.M.)
| | - Enza Lonardo
- Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), Via Pietro Castellino 111, 80131 Naples, Italy;
| |
Collapse
|
42
|
Kennedy CC, Brown EE, Abutaleb NO, Truskey GA. Development and Application of Endothelial Cells Derived From Pluripotent Stem Cells in Microphysiological Systems Models. Front Cardiovasc Med 2021; 8:625016. [PMID: 33659279 PMCID: PMC7917070 DOI: 10.3389/fcvm.2021.625016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/11/2021] [Indexed: 12/02/2022] Open
Abstract
The vascular endothelium is present in all organs and blood vessels, facilitates the exchange of nutrients and waste throughout different organ systems in the body, and sets the tone for healthy vessel function. Mechanosensitive in nature, the endothelium responds to the magnitude and temporal waveform of shear stress in the vessels. Endothelial dysfunction can lead to atherosclerosis and other diseases. Modeling endothelial function and dysfunction in organ systems in vitro, such as the blood-brain barrier and tissue-engineered blood vessels, requires sourcing endothelial cells (ECs) for these biomedical engineering applications. It can be difficult to source primary, easily renewable ECs that possess the function or dysfunction in question. In contrast, human pluripotent stem cells (hPSCs) can be sourced from donors of interest and renewed almost indefinitely. In this review, we highlight how knowledge of vascular EC development in vivo is used to differentiate induced pluripotent stem cells (iPSC) into ECs. We then describe how iPSC-derived ECs are being used currently in in vitro models of organ function and disease and in vivo applications.
Collapse
Affiliation(s)
- Crystal C. Kennedy
- University Program in Genetics and Genomics, Duke University, Durham, NC, United States
| | - Erin E. Brown
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nadia O. Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
43
|
Yoon PS, Del Piccolo N, Shirure VS, Peng Y, Kirane A, Canter RJ, Fields RC, George SC, Gholami S. Advances in Modeling the Immune Microenvironment of Colorectal Cancer. Front Immunol 2021; 11:614300. [PMID: 33643296 PMCID: PMC7902698 DOI: 10.3389/fimmu.2020.614300] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and second leading cause of cancer-related death in the US. CRC frequently metastasizes to the liver and these patients have a particularly poor prognosis. The infiltration of immune cells into CRC tumors and liver metastases accurately predicts disease progression and patient survival. Despite the evident influence of immune cells in the CRC tumor microenvironment (TME), efforts to identify immunotherapies for CRC patients have been limited. Here, we argue that preclinical model systems that recapitulate key features of the tumor microenvironment-including tumor, stromal, and immune cells; the extracellular matrix; and the vasculature-are crucial for studies of immunity in the CRC TME and the utility of immunotherapies for CRC patients. We briefly review the discoveries, advantages, and disadvantages of current in vitro and in vivo model systems, including 2D cell culture models, 3D culture systems, murine models, and organ-on-a-chip technologies.
Collapse
Affiliation(s)
- Paul Sukwoo Yoon
- Department of Surgery, University of California, Davis, Sacramento, CA, United States
| | - Nuala Del Piccolo
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Yushuan Peng
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Amanda Kirane
- Department of Surgery, University of California, Davis, Sacramento, CA, United States
| | - Robert J Canter
- Department of Surgery, University of California, Davis, Sacramento, CA, United States
| | - Ryan C Fields
- Department of Surgery, The Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Sepideh Gholami
- Department of Surgery, University of California, Davis, Sacramento, CA, United States
| |
Collapse
|