1
|
Kataki AD, Gupta PG, Cheema U, Nisbet A, Wang Y, Kocher HM, Pérez-Mancera PA, Velliou EG. Mapping Tumor-Stroma-ECM Interactions in Spatially Advanced 3D Models of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16708-16724. [PMID: 40052705 PMCID: PMC11931495 DOI: 10.1021/acsami.5c02296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/21/2025]
Abstract
Bioengineering-based in vitro tumor models are increasingly important as tools for studying disease progression and therapy response for many cancers, including the deadly pancreatic ductal adenocarcinoma (PDAC) that exhibits a tumor/tissue microenvironment of high cellular/biochemical complexity. Therefore, it is crucial for in vitro models to capture that complexity and to enable investigation of the interplay between cancer cells and factors such as extracellular matrix (ECM) proteins or stroma cells. Using polyurethane (PU) scaffolds, we performed a systematic study on how different ECM protein scaffold coatings impact the long-term cell evolution in scaffolds containing only cancer or only stroma cells (activated stellate and endothelial cells). To investigate potential further changes in those biomarkers due to cancer-stroma interactions, we mapped their expression in dual/zonal scaffolds consisting of a cancer core and a stroma periphery, spatially mimicking the fibrotic/desmoplastic reaction in PDAC. In our single scaffolds, we observed that the protein coating affected the cancer cell spatial aggregation, matrix deposition, and biomarker upregulation in a cell-line-dependent manner. In single stroma scaffolds, different levels of fibrosis/desmoplasia in terms of ECM composition/quantity were generated depending on the ECM coating. When studying the evolution of cancer and stroma cells in our dual/zonal model, biomarkers linked to cell aggressiveness/invasiveness were further upregulated by both cancer and stroma cells as compared to single scaffold models. Collectively, our study advances the understanding of how different ECM proteins impact the long-term cell evolution in PU scaffolds. Our findings show that within our bioengineered models, we can stimulate the cells of the PDAC microenvironment to develop different levels of aggressiveness/invasiveness, as well as different levels of fibrosis. Furthermore, we highlight the importance of considering spatial complexity to map cell invasion. Our work contributes to the design of in vitro models with variable, yet biomimetic, tissue-like properties for studying the tumor microenvironment's role in cancer progression.
Collapse
Affiliation(s)
- Anna-Dimitra Kataki
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Priyanka G. Gupta
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
- School
of Life and Health Sciences, Whitelands College, University of Roehampton, London SW15 4JD, U.K.
| | - Umber Cheema
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Andrew Nisbet
- Department
of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, U.K.
| | - Yaohe Wang
- Centre
for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Hemant M. Kocher
- Centre
for Tumour Biology and Experimental Cancer Medicine, Barts Cancer
Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Pedro A. Pérez-Mancera
- Department
of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Eirini G. Velliou
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| |
Collapse
|
2
|
Garg S, Singla P, Kaur S, Canfarotta F, Velliou E, Dawson JA, Kapur N, Warren NJ, Amarnath S, Peeters M. Future Perspectives on the Automation and Biocompatibility of Molecularly Imprinted Polymers for Healthcare Applications. Macromolecules 2025; 58:1157-1168. [PMID: 39958488 PMCID: PMC11823616 DOI: 10.1021/acs.macromol.4c01621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 02/18/2025]
Abstract
Molecular recognition is of crucial importance in several healthcare applications, such as sensing, drug delivery, and therapeutics. Molecularly imprinted polymers (MIPs) present an interesting alternative to biological receptors (e.g., antibodies, enzymes) for this purpose since synthetic receptors overcome the limited robustness, flexibility, high-cost, and potential for inhibition that comes with natural recognition elements. However, off the shelf MIP products remain limited, which is likely due to the lack of a scalable production approach that can manufacture these materials in high yields and narrow and defined size distributions to have full control over their properties. In this Perspective, we will confer how breakthroughs in the automation of MIP design, manufacturing, and evaluation of performance will accelerate the (commercial) implementation of MIPs in healthcare technology. In addition, we will discuss how prediction of the in vivo behavior of MIPs with animal-free technologies (e.g., 3D tissue models) will be critical to assess their clinical potential.
Collapse
Affiliation(s)
- Saweta Garg
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Pankaj Singla
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| | - Sarbjeet Kaur
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Francesco Canfarotta
- MIP
Discovery, Colworth Park, Sharnbrook, MK44 1LQ, Bedfordshire, United Kingdom
| | - Eirini Velliou
- University
College London, Centre for 3D Models
of Health and Disease, Charles Bell House, London, W1W 7TY, United Kingdom
| | - James A. Dawson
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Nikil Kapur
- University
of Leeds, School of Mechanical Engineering, Woodhouse Lane, Leeds, LS2 9JT, United Kingdom
| | - Nicholas J. Warren
- School of
Chemical, Materials and Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, United Kingdom
| | - Shoba Amarnath
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Marloes Peeters
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| |
Collapse
|
3
|
Akolawala Q, Accardo A. Engineered Cell Microenvironments: A Benchmark Tool for Radiobiology. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5563-5577. [PMID: 39813590 PMCID: PMC11788991 DOI: 10.1021/acsami.4c20455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
The development of engineered cell microenvironments for fundamental cell mechanobiology, in vitro disease modeling, and tissue engineering applications increased exponentially during the last two decades. In such context, in vitro radiobiology is a field of research aiming at understanding the effects of ionizing radiation (e.g., X-rays/photons, high-speed electrons, and high-speed protons) on biological (cancerous) tissues and cells, in particular in terms of DNA damage leading to cell death. Herein, the perspective provides a comparative assessment overview of scaffold-free, scaffold-based, and organ-on-a-chip models for radiobiology, highlighting opportunities, limitations, and future pathways to improve the currently existing approaches toward personalized cancer medicine.
Collapse
Affiliation(s)
- Qais Akolawala
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The
Netherlands
- Holland
Proton Therapy Center (HollandPTC), Huismansingel 4, 2629 JH Delft, The Netherlands
| | - Angelo Accardo
- Department
of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, The
Netherlands
| |
Collapse
|
4
|
Lee Y, Min J, Kim S, Park W, Ko J, Jeon NL. Recapitulating the Cancer-Immunity Cycle on a Chip. Adv Healthc Mater 2025; 14:e2401927. [PMID: 39221688 DOI: 10.1002/adhm.202401927] [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: 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
|
5
|
Acimovic I, Gabrielová V, Martínková S, Eid M, Vlažný J, Moravčík P, Hlavsa J, Moráň L, Cakmakci RC, Staňo P, Procházka V, Kala Z, Trnka J, Vaňhara P. Ex-Vivo 3D Cellular Models of Pancreatic Ductal Adenocarcinoma: From Embryonic Development to Precision Oncology. Pancreas 2025; 54:e57-e71. [PMID: 39074056 DOI: 10.1097/mpa.0000000000002393] [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] [Indexed: 07/31/2024]
Abstract
ABSTRACT Pancreas is a vital gland of gastrointestinal system with exocrine and endocrine secretory functions, interweaved into essential metabolic circuitries of the human body. Pancreatic ductal adenocarcinoma (PDAC) represents one of the most lethal malignancies, with a 5-year survival rate of 11%. This poor prognosis is primarily attributed to the absence of early symptoms, rapid metastatic dissemination, and the limited efficacy of current therapeutic interventions. Despite recent advancements in understanding the etiopathogenesis and treatment of PDAC, there remains a pressing need for improved individualized models, identification of novel molecular targets, and development of unbiased predictors of disease progression. Here we aim to explore the concept of precision medicine utilizing 3-dimensional, patient-specific cellular models of pancreatic tumors and discuss their potential applications in uncovering novel druggable molecular targets and predicting clinical parameters for individual patients.
Collapse
Affiliation(s)
- Ivana Acimovic
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Viktorie Gabrielová
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Stanislava Martínková
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | - Michal Eid
- Departments of Internal Medicine, Hematology and Oncology
| | | | - Petr Moravčík
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Hlavsa
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | | | - Riza Can Cakmakci
- From the Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno
| | - Peter Staňo
- Departments of Internal Medicine, Hematology and Oncology
| | - Vladimír Procházka
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Zdeněk Kala
- Surgery Clinic, University Hospital Brno, Faculty of Medicine, Masaryk University
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague
| | | |
Collapse
|
6
|
Goluba K, Parfejevs V, Rostoka E, Jekabsons K, Blake I, Neimane A, Ule AA, Rimsa R, Vangravs R, Pcolkins A, Riekstina U. Personalized PDAC chip with functional endothelial barrier for tumour biomarker detection: A platform for precision medicine applications. Mater Today Bio 2024; 29:101262. [PMID: 39381267 PMCID: PMC11460472 DOI: 10.1016/j.mtbio.2024.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterised by poor survival rates and an increasing global incidence. Advances in the staging and categorization of pancreatic tumours, along with the discovery of functional mutations, have made precision treatments possible, which may lead to better clinical results. To further improve customized treatment approaches, in vitro models that can be used for functional drug sensitivity testing and precisely mimic the disease at the organ level are required. In this study, we present a workflow for creating a personalized PDAC chip utilising primary tumour-derived human pancreatic organoids (hPOs) and Human Umbilical Vein Endothelial Cells (HUVECs) to simulate the vascular barrier and tumour interactions within a PDMS-free organ-on-a-chip system. The patient PDAC tissue, expanded as tumour hPOs, could be cultured as adherent cells on the chip for more than 50 days, allowing continuous monitoring of cell viability through outflows from tumour and endothelial channels. Our findings demonstrate a gradual increase in cell density and cell turnover in the pancreatic tumor channel. Tumour-specific biomarkers, including CA-19.9, TIMP-1, Osteopontin, MIC-1, ICAM-1 and sAXL were consistently detected in the PDAC chip outflows. Comparative analyses between tissue culture plates and microfluidic conditions revealed significant differences in biomarker secretion patterns, highlighting the advantages of the microfluidics approach. This PDAC chip provides a stable, reproducible tumour model system with a functional endothelial cell barrier, suitable for drug sensitivity and secretory biomarker studies, thus serving as a platform for functional precision medicine application and multi-organ chip development.
Collapse
Affiliation(s)
- Karina Goluba
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Vadims Parfejevs
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Evita Rostoka
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Kaspars Jekabsons
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Ilze Blake
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Anastasija Neimane
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| | - Annija Anete Ule
- Institute of Solid State Physics, University of Latvia, Kengaraga iela 8, Riga, Latvia
| | - Roberts Rimsa
- Institute of Solid State Physics, University of Latvia, Kengaraga iela 8, Riga, Latvia
| | - Reinis Vangravs
- Latvian Centre of Infectious Diseases, Laboratory Service, Riga East University Hospital, Linezera iela 3, LV-1006, Riga, Latvia
| | - Andrejs Pcolkins
- Department of Abdominal and Soft Tissue Surgery, Riga East Clinical University Hospital, Hipokrata iela 2, Riga, Latvia
| | - Una Riekstina
- Pharmaceutical Sciences Center, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Riga, Latvia
| |
Collapse
|
7
|
Yadav P, Singh S, Jaiswal S, Kumar R. Synthetic and natural polymer hydrogels: A review of 3D spheroids and drug delivery. Int J Biol Macromol 2024; 280:136126. [PMID: 39349080 DOI: 10.1016/j.ijbiomac.2024.136126] [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: 04/22/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
This review centers on the synthesis and characterization of both natural and synthetic hydrogels, highlighting their diverse applications across various fields. We will delve into the evolution of hydrogels, focusing on the importance of polysaccharide-based and synthetic variants, which have been particularly chosen for 3D spheroid development in cancer research and drug delivery. A detailed background on the research and specific methodologies, including the in-situ free radical polymerization used for synthesizing these hydrogels, will be extensively discussed. Additionally, the review will explore various applications of these hydrogels, such as their self-healing properties, swelling ratios, pH responsiveness, and cell viability. A comprehensive literature review will support this investigation. Ultimately, this review aims to clearly outline the objectives and significance of hydrogel synthesis and their applications.
Collapse
Affiliation(s)
- Paramjeet Yadav
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Shiwani Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Sheetal Jaiswal
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India
| | - Rajesh Kumar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, UP, India.
| |
Collapse
|
8
|
Singla P, Broughton T, Sullivan MV, Garg S, Berlinguer‐Palmini R, Gupta P, Smith KJ, Gardner B, Canfarotta F, Turner NW, Velliou E, Amarnath S, Peeters M. Double Imprinted Nanoparticles for Sequential Membrane-to-Nuclear Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309976. [PMID: 38973256 PMCID: PMC11423068 DOI: 10.1002/advs.202309976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Efficient and site-specific delivery of therapeutics drugs remains a critical challenge in cancer treatment. Traditional drug nanocarriers such as antibody-drug conjugates are not generally accessible due to their high cost and can lead to serious side effects including life-threatening allergic reactions. Here, these problems are overcome via the engineering of supramolecular agents that are manufactured with an innovative double imprinting approach. The developed molecularly imprinted nanoparticles (nanoMIPs) are targeted toward a linear epitope of estrogen receptor alfa (ERα) and loaded with the chemotherapeutic drug doxorubicin. These nanoMIPs are cost-effective and rival the affinity of commercial antibodies for ERα. Upon specific binding of the materials to ERα, which is overexpressed in most breast cancers (BCs), nuclear drug delivery is achieved via receptor-mediated endocytosis. Consequentially, significantly enhanced cytotoxicity is elicited in BC cell lines overexpressing ERα, paving the way for precision treatment of BC. Proof-of-concept for the clinical use of the nanoMIPs is provided by evaluating their drug efficacy in sophisticated three-dimensional (3D) cancer models, which capture the complexity of the tumor microenvironment in vivo without requiring animal models. Thus, these findings highlight the potential of nanoMIPs as a promising class of novel drug compounds for use in cancer treatment.
Collapse
Affiliation(s)
- Pankaj Singla
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Thomas Broughton
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- NIHR, Biomedical Research CentreNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Mark V. Sullivan
- Department of ChemistryUniversity of SheffieldDainton BuildingSheffieldS3 7HFUK
| | - Saweta Garg
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Rolando Berlinguer‐Palmini
- The Bio‐Imaging Unit, Medical SchoolNewcastle UniversityWilliam Leech BuildingNewcastle Upon TyneNE2 4HHUK
| | - Priyanka Gupta
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional ScienceUniversity College LondonLondonW1W 7TYUK
| | - Katie J Smith
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Ben Gardner
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | | | - Nicholas W. Turner
- Department of ChemistryUniversity of SheffieldDainton BuildingSheffieldS3 7HFUK
| | - Eirini Velliou
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional ScienceUniversity College LondonLondonW1W 7TYUK
| | - Shoba Amarnath
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- NIHR, Biomedical Research CentreNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Marloes Peeters
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| |
Collapse
|
9
|
Sgarminato V, Madrid-Wolff J, Boniface A, Ciardelli G, Tonda-Turo C, Moser C. 3D in vitromodeling of the exocrine pancreatic unit using tomographic volumetric bioprinting. Biofabrication 2024; 16:045034. [PMID: 39121863 DOI: 10.1088/1758-5090/ad6d8d] [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: 04/03/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevantin vitrohuman models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.
Collapse
Affiliation(s)
- Viola Sgarminato
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Jorge Madrid-Wolff
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Antoine Boniface
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Christophe Moser
- Laboratory of Applied Photonics Devices, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Gupta P, Bermejo-Rodriguez C, Kocher H, Pérez-Mancera PA, Velliou EG. Chemotherapy Assessment in Advanced Multicellular 3D Models of Pancreatic Cancer: Unravelling the Importance of Spatiotemporal Mimicry of the Tumor Microenvironment. Adv Biol (Weinh) 2024; 8:e2300580. [PMID: 38327154 DOI: 10.1002/adbi.202300580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenge for global health with very low survival rate and high therapeutic resistance. Hence, advanced preclinical models for treatment screening are of paramount importance. Herein, chemotherapeutic (gemcitabine) assessment on novel (polyurethane) scaffold-based spatially advanced 3D multicellular PDAC models is carried out. Through comprehensive image-based analysis at the protein level, and expression analysis at the mRNA level, the importance of stromal cells is confirmed, primarily activated stellate cells in the chemoresistance of PDAC cells within the models. Furthermore, it is demonstrated that, in addition to the presence of activated stellate cells, the spatial architecture of the scaffolds, i.e., segregation/compartmentalization of the cancer and stromal zones, affect the cellular evolution and is necessary for the development of chemoresistance. These results highlight that, further to multicellularity, mapping the tumor structure/architecture and zonal complexity in 3D cancer models is important for better mimicry of the in vivo therapeutic response.
Collapse
Affiliation(s)
- Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, W1W 7TY, UK
| | - Camino Bermejo-Rodriguez
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Hemant Kocher
- Centre for Tumour Biology and Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Pedro A Pérez-Mancera
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Eirini G Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, W1W 7TY, UK
| |
Collapse
|
11
|
Angelopoulou A. Nanostructured Biomaterials in 3D Tumor Tissue Engineering Scaffolds: Regenerative Medicine and Immunotherapies. Int J Mol Sci 2024; 25:5414. [PMID: 38791452 PMCID: PMC11121067 DOI: 10.3390/ijms25105414] [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: 04/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The evaluation of nanostructured biomaterials and medicines is associated with 2D cultures that provide insight into biological mechanisms at the molecular level, while critical aspects of the tumor microenvironment (TME) are provided by the study of animal xenograft models. More realistic models that can histologically reproduce human tumors are provided by tissue engineering methods of co-culturing cells of varied phenotypes to provide 3D tumor spheroids that recapitulate the dynamic TME in 3D matrices. The novel approaches of creating 3D tumor models are combined with tumor tissue engineering (TTE) scaffolds including hydrogels, bioprinted materials, decellularized tissues, fibrous and nanostructured matrices. This review focuses on the use of nanostructured materials in cancer therapy and regeneration, and the development of realistic models for studying TME molecular and immune characteristics. Tissue regeneration is an important aspect of TTE scaffolds used for restoring the normal function of the tissues, while providing cancer treatment. Thus, this article reports recent advancements in the development of 3D TTE models for antitumor drug screening, studying tumor metastasis, and tissue regeneration. Also, this review identifies the significant opportunities of using 3D TTE scaffolds in the evaluation of the immunological mechanisms and processes involved in the application of immunotherapies.
Collapse
Affiliation(s)
- Athina Angelopoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
| |
Collapse
|
12
|
Howard CJ, Paul A, Duruanyanwu J, Sackho K, Campagnolo P, Stolojan V. The Manufacturing Conditions for the Direct and Reproducible Formation of Electrospun PCL/Gelatine 3D Structures for Tissue Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3107. [PMID: 38133004 PMCID: PMC10745430 DOI: 10.3390/nano13243107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
Electrospinning is a versatile technique for fabricating nanofibrous scaffolds for tissue engineering applications. However, the direct formation of 3D sponges through electrospinning has previously not been reproducible. We used a Taguchi experimental design approach to optimise the electrospinning parameters for forming PCL and PCL/gelatine 3D sponges. The following parameters were investigated to improve sponge formation: solution concentration, humidity, and solution conductivity. Pure PCL sponges were achievable. However, a much fluffier sponge formed by increasing the solution conductivity with gelatine. The optimal conditions for sponge formation 24 w/v% 80:20 PCL:gelatine on aluminium foil at ≥70% humidity, 15 cm, 22 kV and 1500 µL/h. The resulting sponge had a highly porous structure with a fibre diameter of ~1 µm. They also supported significantly higher cell viability than 2D electrospun mats, dropcast films of the same material and even the TCP positive control. Our study demonstrates that the direct formation of PCL/gelatine 3D sponges through electrospinning is feasible and promising for tissue engineering applications. The sponges have a highly porous structure and support cell viability, which are essential properties for tissue engineering scaffolds. Further studies are needed to optimise the manufacturing process and evaluate the sponges' long-term performance in vivo.
Collapse
Affiliation(s)
- Chloe Jayne Howard
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| | - Aumrita Paul
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| | - Justin Duruanyanwu
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Kenza Sackho
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Paola Campagnolo
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Vlad Stolojan
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| |
Collapse
|
13
|
Dellaquila A, Dujardin C, Le Bao C, Chaumeton C, Carré A, Le Guilcher C, Lam F, Simon-Yarza T. Fibroblasts mediate endothelium response to angiogenic cues in a newly developed 3D stroma engineered model. BIOMATERIALS ADVANCES 2023; 154:213636. [PMID: 37778292 DOI: 10.1016/j.bioadv.2023.213636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/30/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Three-dimensional stroma engineered models would enable fundamental and applicative studies of human tissues interaction and remodeling in both physiological and pathological conditions. In this work, we propose a 3D vascularized stroma model to be used as in vitro platform for drug testing. A pullulan/dextran-based porous scaffold containing pre-patterned microchannels of 100 μm diameter is used for co-culturing of fibroblasts within the matrix pores and endothelial cells to form the lumen. Optical clearing of the constructs by hyperhydration allows for in-depth imaging of the model up to 1 mm by lightsheet and confocal microscopy. Our 3D vascularized stroma model allows for higher viability, metabolism and cytokines expression compared to a monocultured vascular model. Stroma-endothelium cross-talk is then investigated by exposing the system to pro and anti-angiogenic molecules. The results highlight the protective role played by fibroblasts on the vasculature, as demonstrated by decreased cytotoxicity, restoration of nitric oxide levels upon challenge, and sustained expression of endothelial markers CD31, vWF and VEGF. Our tissue model provides a 3D engineered platform for in vitro studies of stroma remodeling in angiogenesis-driven events, known to be a leading mechanism in diseased conditions, such as metastatic cancers, retinopathies and ischemia, and to investigate related potential therapies.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France.
| | - Chloé Dujardin
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - Chau Le Bao
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - Chloé Chaumeton
- Sorbonne Université, Institute of Biology Paris-Seine, Paris 75005, France
| | - Albane Carré
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - Camille Le Guilcher
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France
| | - France Lam
- Sorbonne Université, Institute of Biology Paris-Seine, Paris 75005, France
| | - Teresa Simon-Yarza
- Université Paris Cité, Université Sorbonne Paris Nord, Laboratory for Vascular Translational Science, INSERM U1148, X. Bichat Hospital, Paris 75018, France.
| |
Collapse
|
14
|
Sun L, Wang X, He Y, Chen B, Shan B, Yang J, Wang R, Zeng X, Li J, Tan H, Liang R. Polyurethane scaffold-based 3D lung cancer model recapitulates in vivo tumor biological behavior for nanoparticulate drug screening. Regen Biomater 2023; 10:rbad091. [PMID: 37965109 PMCID: PMC10641150 DOI: 10.1093/rb/rbad091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 11/16/2023] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Preclinical studies in lung cancer hold the promise of screening for effective antitumor agents, but mechanistic studies and drug discovery based on 2D cell models have a high failure rate in getting to the clinic. Thus, there is an urgent need to explore more reliable and effective in vitro lung cancer models. Here, we prepared a series of three-dimensional (3D) waterborne biodegradable polyurethane (WBPU) scaffolds as substrates to establish biomimetic tumor models in vitro. These 3D WBPU scaffolds were porous and could absorb large amounts of free water, facilitating the exchange of substances (nutrients and metabolic waste) and cell growth. The scaffolds at wet state could simulate the mechanics (elastic modulus ∼1.9 kPa) and morphology (porous structures) of lung tissue and exhibit good biocompatibility. A549 lung cancer cells showed adherent growth pattern and rapidly formed 3D spheroids on WBPU scaffolds. Our results showed that the scaffold-based 3D lung cancer model promoted the expression of anti-apoptotic and epithelial-mesenchymal transition-related genes, giving it a more moderate growth and adhesion pattern compared to 2D cells. In addition, WBPU scaffold-established 3D lung cancer model revealed a closer expression of proteins to in vivo tumor, including tumor stem cell markers, cell proliferation, apoptosis, invasion and tumor resistance proteins. Based on these features, we further demonstrated that the 3D lung cancer model established by the WBPU scaffold was very similar to the in vivo tumor in terms of both resistance and tolerance to nanoparticulate drugs. Taken together, WBPU scaffold-based lung cancer model could better mimic the growth, microenvironment and drug response of tumor in vivo. This emerging 3D culture system holds promise to shorten the formulation cycle of individualized treatments and reduce the use of animals while providing valid research data for clinical trials.
Collapse
Affiliation(s)
- Lu Sun
- Department of Targeting Therapy & Immunology; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaofei Wang
- Department of Medical Polymer Materials; Department of Artificial Organism, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People’s Republic of China
| | - Yushui He
- Department of Medical Polymer Materials; Department of Artificial Organism, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People’s Republic of China
| | - Boran Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Baoyin Shan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Jinlong Yang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Ruoran Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Xihang Zeng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Jiehua Li
- Department of Medical Polymer Materials; Department of Artificial Organism, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People’s Republic of China
| | - Hong Tan
- Department of Medical Polymer Materials; Department of Artificial Organism, College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People’s Republic of China
| | - Ruichao Liang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| |
Collapse
|
15
|
De Vlieghere E, Van de Vijver K, Blondeel E, Carpentier N, Ghobeira R, Pauwels J, Riemann S, Minsart M, Fieuws C, Mestach J, Baeyens A, De Geyter N, Debbaut C, Denys H, Descamps B, Claes K, Vral A, Van Dorpe J, Gevaert K, De Geest BG, Ceelen W, Van Vlierberghe S, De Wever O. A preclinical platform for assessing long-term drug efficacy exploiting mechanically tunable scaffolds colonized by a three-dimensional tumor microenvironment. Biomater Res 2023; 27:104. [PMID: 37853495 PMCID: PMC10583378 DOI: 10.1186/s40824-023-00441-3] [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: 01/26/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Long-term drug evaluation heavily relies upon rodent models. Drug discovery methods to reduce animal models in oncology may include three-dimensional (3D) cellular systems that take into account tumor microenvironment (TME) cell types and biomechanical properties. METHODS In this study we reconstructed a 3D tumor using an elastic polymer (acrylate-endcapped urethane-based poly(ethylene glycol) (AUPPEG)) with clinical relevant stiffness. Single cell suspensions from low-grade serous ovarian cancer (LGSOC) patient-derived early passage cultures of cancer cells and cancer-associated fibroblasts (CAF) embedded in a collagen gel were introduced to the AUPPEG scaffold. After self-organization in to a 3D tumor, this model was evaluated by a long-term (> 40 days) exposure to a drug combination of MEK and HSP90 inhibitors. The drug-response results from this long-term in vitro model are compared with drug responses in an orthotopic LGSOC xenograft mouse model. RESULTS The in vitro 3D scaffold LGSOC model mimics the growth ratio and spatial organization of the LGSOC. The AUPPEG scaffold approach allows to test new targeted treatments and monitor long-term drug responses. The results correlate with those of the orthotopic LGSOC xenograft mouse model. CONCLUSIONS The mechanically-tunable scaffolds colonized by a three-dimensional LGSOC allow long-term drug evaluation and can be considered as a valid alternative to reduce, replace and refine animal models in drug discovery.
Collapse
Affiliation(s)
- Elly De Vlieghere
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Koen Van de Vijver
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Eva Blondeel
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Nathan Carpentier
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Rouba Ghobeira
- Department of Applied Physics, Research Unit Plasma Technology (RUPT), Ghent University, Ghent, Belgium
| | - Jarne Pauwels
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent University, Ghent, Belgium
| | - Sebastian Riemann
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Manon Minsart
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Charlotte Fieuws
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Johanna Mestach
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Ans Baeyens
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Human Structure and Repair, Radiobiology Group, Ghent University, Ghent, Belgium
| | - Nathalie De Geyter
- Department of Applied Physics, Research Unit Plasma Technology (RUPT), Ghent University, Ghent, Belgium
| | - Charlotte Debbaut
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Electronics and Information Systems, IBiTech-Biommeda, Ghent University, Ghent, Belgium
| | - Hannelore Denys
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Electronics and Information Systems, IbiTech-Medisip, Ghent University, Ghent, Belgium
| | - Kathleen Claes
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Anne Vral
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Human Structure and Repair, Radiobiology Group, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Kris Gevaert
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, VIB Center for Medical Biotechnology, Ghent University, Ghent, Belgium
| | - Bruno G De Geest
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.
- Department of Pharmaceutics, Ghent University, Ghent, Belgium.
| |
Collapse
|
16
|
Saunders KD, von Gerichten J, Lewis HM, Gupta P, Spick M, Costa C, Velliou E, Bailey MJ. Single-Cell Lipidomics Using Analytical Flow LC-MS Characterizes the Response to Chemotherapy in Cultured Pancreatic Cancer Cells. Anal Chem 2023; 95:14727-14735. [PMID: 37725657 PMCID: PMC10551860 DOI: 10.1021/acs.analchem.3c02854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
In this work, we demonstrate the development and first application of nanocapillary sampling followed by analytical flow liquid chromatography-mass spectrometry for single-cell lipidomics. Around 260 lipids were tentatively identified in a single cell, demonstrating remarkable sensitivity. Human pancreatic ductal adenocarcinoma cells (PANC-1) treated with the chemotherapeutic drug gemcitabine can be distinguished from controls solely on the basis of their single-cell lipid profiles. Notably, the relative abundance of LPC(0:0/16:0) was significantly affected in gemcitabine-treated cells, in agreement with previous work in bulk. This work serves as a proof of concept that live cells can be sampled selectively and then characterized using automated and widely available analytical workflows, providing biologically relevant outputs.
Collapse
Affiliation(s)
| | | | - Holly-May Lewis
- Faculty
of Health & Medical Sciences, University
of Surrey, Guildford GU2 7XH, U.K.
| | - Priyanka Gupta
- Centre
for 3D Models of Health and Disease, University
College London—Division of Surgery and Interventional Science, London W1W 7TY, U.K.
| | - Matt Spick
- Faculty
of Health & Medical Sciences, University
of Surrey, Guildford GU2 7XH, U.K.
| | - Catia Costa
- Ion
Beam Centre, University of Surrey, Guildford GU2 7XH, U.K.
| | - Eirini Velliou
- Centre
for 3D Models of Health and Disease, University
College London—Division of Surgery and Interventional Science, London W1W 7TY, U.K.
| | - Melanie J. Bailey
- Department
of Chemistry, University of Surrey, Guildford GU2 7XH, U.K.
| |
Collapse
|
17
|
Urciuolo F, Imparato G, Netti PA. In vitro strategies for mimicking dynamic cell-ECM reciprocity in 3D culture models. Front Bioeng Biotechnol 2023; 11:1197075. [PMID: 37434756 PMCID: PMC10330728 DOI: 10.3389/fbioe.2023.1197075] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
The extracellular microenvironment regulates cell decisions through the accurate presentation at the cell surface of a complex array of biochemical and biophysical signals that are mediated by the structure and composition of the extracellular matrix (ECM). On the one hand, the cells actively remodel the ECM, which on the other hand affects cell functions. This cell-ECM dynamic reciprocity is central in regulating and controlling morphogenetic and histogenetic processes. Misregulation within the extracellular space can cause aberrant bidirectional interactions between cells and ECM, resulting in dysfunctional tissues and pathological states. Therefore, tissue engineering approaches, aiming at reproducing organs and tissues in vitro, should realistically recapitulate the native cell-microenvironment crosstalk that is central for the correct functionality of tissue-engineered constructs. In this review, we will describe the most updated bioengineering approaches to recapitulate the native cell microenvironment and reproduce functional tissues and organs in vitro. We have highlighted the limitations of the use of exogenous scaffolds in recapitulating the regulatory/instructive and signal repository role of the native cell microenvironment. By contrast, strategies to reproduce human tissues and organs by inducing cells to synthetize their own ECM acting as a provisional scaffold to control and guide further tissue development and maturation hold the potential to allow the engineering of fully functional histologically competent three-dimensional (3D) tissues.
Collapse
Affiliation(s)
- F. Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - G. Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - P. A. Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|
18
|
Wishart G, Gupta P, Nisbet A, Velliou E, Schettino G. Enhanced effect of X-rays in the presence of a static magnetic field within a 3D pancreatic cancer model. Br J Radiol 2023; 96:20220832. [PMID: 36475863 PMCID: PMC9975369 DOI: 10.1259/bjr.20220832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To evaluate the impact of static magnetic field (SMF) presence on the radiation response of pancreatic cancer cells in polyurethane-based highly macro-porous scaffolds in hypoxic (1% O2) and normoxic (21% O2) conditions, towards understanding MR-guided radiotherapy, shedding light on the potential interaction phenomenon between SMF and radiation in a three-dimensional (3D) microenvironment. METHODS Pancreatic cancer cells (PANC-1, ASPC-1) were seeded into fibronectin-coated highly porous polyethene scaffolds for biomimicry and cultured for 4 weeks in in vitro normoxia (21% O2) followed by a 2-day exposure to either in vitro hypoxia (1% O2) or maintenance in in vitro normoxia (21% O2). The samples were then irradiated with 6 MV photons in the presence or absence of a 1.5 T field. Thereafter, in situ post-radiation monitoring (1 and 7 days post-irradiation treatment) took place via quantification of (i) live dead and (ii) apoptotic profiles. RESULTS We report: (i) pancreatic ductal adenocarcinoma hypoxia-associated radioprotection, in line with our previous findings, (ii) an enhanced effect of radiation in the presence of SMFin in vitro hypoxia (1% O2) for both short- (1 day) and long-term (7 days) post -radiation analysis and (iii) an enhanced effect of radiation in the presence of SMF in in vitro normoxia (21% O2) for long-term (7 days) post-radiation analysis within a 3D pancreatic cancer model. CONCLUSION With limited understanding of the potential interaction phenomenon between SMF and radiation, this 3D system allows combination evaluation for a cancer in which the role of radiotherapy is still evolving. ADVANCES IN KNOWLEDGE This study examined the use of a 3D model to investigate MR-guided radiotherapy in a hypoxic microenvironment, indicating that this could be a useful platform to further understanding of SMF influence on radiation.
Collapse
Affiliation(s)
| | | | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London (UCL), London, UK
| | | | | |
Collapse
|
19
|
Gupta P, Velliou EG. A Step-by-Step Methodological Guide for Developing Zonal Multicellular Scaffold-Based Pancreatic Cancer Models. Methods Mol Biol 2023; 2645:221-229. [PMID: 37202622 DOI: 10.1007/978-1-0716-3056-3_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The tumor microenvironment (TME), a complex heterogeneous mixture of various cellular, physical, and biochemical components and signals, is a major player in the process of tumor growth and its response to therapeutic methods. In vitro 2D monocellular cancer models are unable to mimic the complex in vivo characteristics of cancer TME involving cellular heterogeneity, presence of extracellular matrix (ECM) proteins, as well as spatial orientation and organization of different cell types forming the TME. In vivo animal-based studies have ethical concerns, are expensive and time-consuming, and involve models of non-human species. In vitro 3D models are capable of tiding over several issues associated with both 2D in vitro and in vivo animal models. We have recently developed a novel zonal multicellular 3D in vitro model for pancreatic cancer involving cancer cells, endothelial cells, and pancreatic stellate cells. Our model (i) can provide long-term culture (up to 4 weeks), (ii) can control the ECM biochemical configuration in a cell specific manner, (iii) shows large amounts of collagen secretion by the stellate cells mimicking desmoplasia, and (iv) expresses cell-specific markers throughout the whole culture period. This chapter describes the experimental methodology to form our hybrid multicellular 3D model for pancreatic ductal adenocarcinoma, including the immunofluorescence staining on the cell culture.
Collapse
Affiliation(s)
- Priyanka Gupta
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, UK
| | - Eirini G Velliou
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, UK.
| |
Collapse
|
20
|
Imparato G, Urciuolo F, Mazio C, Netti PA. Capturing the spatial and temporal dynamics of tumor stroma for on-chip optimization of microenvironmental targeting nanomedicine. LAB ON A CHIP 2022; 23:25-43. [PMID: 36305728 DOI: 10.1039/d2lc00611a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Malignant cells grow in a complex microenvironment that plays a key role in cancer progression. The "dynamic reciprocity" existing between cancer cells and their microenvironment is involved in cancer differentiation, proliferation, invasion, metastasis, and drug response. Therefore, understanding the molecular mechanisms underlying the crosstalk between cancer cells and their surrounding tissue (i.e., tumor stroma) and how this interplay affects the disease progression is fundamental to design and validate novel nanotherapeutic approaches. As an important regulator of tumor progression, metastasis and therapy resistance, the extracellular matrix of tumors, the acellular component of the tumor microenvironment, has been identified as very promising target of anticancer treatment, revolutionizing the traditional therapeutic paradigm that sees the neoplastic cells as the preferential objective to fight cancer. To design and to validate such a target therapy, advanced 3D preclinical models are necessary to correctly mimic the complex, dynamic and heterogeneous tumor microenvironment. In addition, the recent advancement in microfluidic technology allows fine-tuning and controlling microenvironmental parameters in tissue-on-chip devices in order to emulate the in vivo conditions. In this review, after a brief description of the origin of tumor microenvironment heterogeneity, some examples of nanomedicine approaches targeting the tumor microenvironment have been reported. Further, how advanced 3D bioengineered tumor models coupled with a microfluidic device can improve the design and testing of anti-cancer nanomedicine targeting the tumor microenvironment has been discussed. We highlight that the presence of a dynamic extracellular matrix, able to capture the spatiotemporal heterogeneity of tumor stroma, is an indispensable requisite for tumor-on-chip model and nanomedicine testing.
Collapse
Affiliation(s)
- Giorgia Imparato
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| | - Claudia Mazio
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
| | - Paolo A Netti
- Center for Advanced Biomaterials for Health Care@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci n. 53, 80125 Napoli, Italy.
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, 80125 Napoli, Italy
| |
Collapse
|
21
|
Xie D, Jia S, Ping D, Wang D, Cao L. Scaffold-based three-dimensional cell model of pancreatic cancer is more suitable than scaffold-free three-dimensional cell model of pancreatic cancer for drug discovery. Cytotechnology 2022; 74:657-667. [PMID: 36389286 PMCID: PMC9652184 DOI: 10.1007/s10616-022-00553-z] [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] [Received: 10/29/2021] [Accepted: 10/03/2022] [Indexed: 11/03/2022] Open
Abstract
Pancreatic cancer is one of the deadliest malignancies. Three-dimensional (3D) pancreatic cancer cell models for drug screening have been established to improve treatment for pancreatic cancer. However, few studies focus on different drug responses and drug-related molecular mechanisms in various types of 3D cell models. In this study, we constructed 3D scaffold-free cell models and 3D scaffold-based cell models of pancreatic cancer, evaluated chemotherapeutic drug responses in different 3D models, assessed clinical relevance of the models, and investigated molecular mechanisms of chemoresistance and drug pathways in different 3D models. Both types of 3D models showed resistance to chemotherapeutic drugs, and scaffold-based pancreatic cancer models could better reflect in vivo drug efficacy than 2D and scaffold-free pancreatic cancer models did. Increased cell adhesion, extracellular matrix (ECM) synthesis and drug transport were essential for drug resistance in 3D models, and anti-apoptosis might contribute to extreme chemoresistance in scaffold-free models. Moreover, scaffold-based pancreatic cancer models were more suitable than scaffold-free models for drug pathway research.
Collapse
Affiliation(s)
- Dafei Xie
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
- Department of General Surgery, Zhejiang Hospital, Hangzhou, 310000 China
| | - Shengnan Jia
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Dongnan Ping
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Dong Wang
- Department of General Surgery, Zhejiang Hospital, Hangzhou, 310000 China
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| |
Collapse
|
22
|
Sevinyan L, Gupta P, Velliou E, Madhuri TK. The Development of a Three-Dimensional Platform for Patient-Derived Ovarian Cancer Tissue Models: A Systematic Literature Review. Cancers (Basel) 2022; 14:5628. [PMID: 36428724 PMCID: PMC9688222 DOI: 10.3390/cancers14225628] [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: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
There is an unmet biomedical need for ex vivo tumour models that would predict drug responses and in turn help determine treatment regimens and potentially predict resistance before clinical studies. Research has shown that three dimensional models of ovarian cancer (OvCa) are more realistic than two dimensional in vitro systems as they are able to capture patient in vivo conditions in more accurate manner. The vast majority of studies aiming to recapitulate the ovarian tumour morphology, behaviors, and study chemotherapy responses have been using ovarian cancer cell lines. However, despite the advantages of utilising cancer cell lines to set up a platform, they are not as informative as systems applying patient derived cells, as cell lines are not able to recapitulate differences between each individual patient characteristics. In this review we discussed the most recent advances in the creation of 3D ovarian cancer models that have used patient derived material, the challenges to overcome and future applications.
Collapse
Affiliation(s)
- Lusine Sevinyan
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| | - Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Thumuluru Kavitha Madhuri
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| |
Collapse
|
23
|
Esmaeili J, Barati A, Charelli LE. Discussing the final size and shape of the reconstructed tissues in tissue engineering. J Artif Organs 2022:10.1007/s10047-022-01360-1. [PMID: 36125581 DOI: 10.1007/s10047-022-01360-1] [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: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
Tissue engineering (TE) has made a revolution in repairing, replacing, or regenerating tissues or organs, but it has still a long way ahead. The mechanical properties along with suitable physicochemical and biological characteristics are the initial criteria for scaffolds in TE that should be fulfilled. This research will provide another point of view toward TE challenges concerning the morphological and geometrical aspects of the reconstructed tissue and which parameters may affect it. Based on our survey, there is a high possibility that the final reconstructed tissue may be different in size and shape compared to the original design scaffold. Thereby, the 3D-printed scaffold might not guarantee an accurate tissue reconstruction. The main justification for this is the unpredicted behavior of cells, specifically in the outer layer of the scaffold. It can also be a concern when the scaffold is implanted while cell migration cannot be controlled through the in vivo signaling pathways, which might cause cancer challenges. To sum up, it is concluded that more studies are necessary to focus on the size and geometry of the final reconstructed tissue.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.,Tissue Engineering Department, TISSUEHUB Co., Tehran, Iran
| | - Aboulfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.
| | - Letícia Emiliano Charelli
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Rengaraj A, Bosc L, Machillot P, McGuckin C, Milet C, Forraz N, Paliard P, Barbier D, Picart C. Engineering of a Microscale Niche for Pancreatic Tumor Cells Using Bioactive Film Coatings Combined with 3D-Architectured Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13107-13121. [PMID: 35275488 PMCID: PMC7614000 DOI: 10.1021/acsami.2c01747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Two-photon polymerization has recently emerged as a promising technique to fabricate scaffolds for three-dimensional (3D) cell culture and tissue engineering. Here, we combined 3D-printed microscale scaffolds fabricated using two-photon polymerization with a bioactive layer-by-layer film coating. This bioactive coating consists of hyaluronic acid and poly(l-lysine) of controlled stiffness, loaded with fibronectin and bone morphogenic proteins 2 and 4 (BMP2 and BMP4) as matrix-bound proteins. Planar films were prepared using a liquid handling robot directly in 96-well plates to perform high-content studies of cellular processes, especially cell adhesion, proliferation, and BMP-induced signaling. The behaviors of two human pancreatic cell lines PANC1 (immortalized) and PAN092 (patient-derived cell line) were systematically compared and revealed important context-specific cell responses, notably in response to film stiffness and matrix-bound BMPs (bBMPs). Fibronectin significantly increased cell adhesion, spreading, and proliferation for both cell types on soft and stiff films; BMP2 increased cell adhesion and inhibited proliferation of PANC1 cells and PAN092 on soft films. BMP4 enhanced cell adhesion and proliferation of PANC1 and showed a bipolar effect on PAN092. Importantly, PANC1 exhibited a strong dose-dependent BMP response, notably for bBMP2, while PAN092 was insensitive to BMPs. Finally, we proved that it is possible to combine a microscale 3D Ormocomp scaffold fabricated using the two-photon polymerization technique with the bioactive film coating to form a microscale tumor tissue and mimic the early stages of metastatic cancer.
Collapse
Affiliation(s)
- Arunkumar Rengaraj
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Lauriane Bosc
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Colin McGuckin
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Clément Milet
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Nico Forraz
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Philippe Paliard
- Microlight 3D, 5 avenue du Grand Sablon, 38700 La Tronche, France
| | - Denis Barbier
- Microlight 3D, 5 avenue du Grand Sablon, 38700 La Tronche, France
| | - Catherine Picart
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
- Institut Universitaire de France (IUF), Ministère de l’Enseignement Supérieur, de la Recherche et de I’Industrie, 1 rue Descartes, 75 231 Paris Cedex 05, France
| |
Collapse
|
25
|
Mastrullo V, van der Veen DR, Gupta P, Matos RS, Johnston JD, McVey JH, Madeddu P, Velliou EG, Campagnolo P. Pericytes' Circadian Clock Affects Endothelial Cells' Synchronization and Angiogenesis in a 3D Tissue Engineered Scaffold. Front Pharmacol 2022; 13:867070. [PMID: 35387328 PMCID: PMC8977840 DOI: 10.3389/fphar.2022.867070] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/23/2022] [Indexed: 01/02/2023] Open
Abstract
Angiogenesis, the formation of new capillaries from existing ones, is a fundamental process in regenerative medicine and tissue engineering. While it is known to be affected by circadian rhythms in vivo, its peripheral regulation within the vasculature and the role it performs in regulating the interplay between vascular cells have not yet been investigated. Peripheral clocks within the vasculature have been described in the endothelium and in smooth muscle cells. However, to date, scarce evidence has been presented regarding pericytes, a perivascular cell population deeply involved in the regulation of angiogenesis and vessel maturation, as well as endothelial function and homeostasis. More crucially, pericytes are also a promising source of cells for cell therapy and tissue engineering. Here, we established that human primary pericytes express key circadian genes and proteins in a rhythmic fashion upon synchronization. Conversely, we did not detect the same patterns in cultured endothelial cells. In line with these results, pericytes' viability was disproportionately affected by circadian cycle disruption, as compared to endothelial cells. Interestingly, endothelial cells' rhythm could be induced following exposure to synchronized pericytes in a contact co-culture. We propose that this mechanism could be linked to the altered release/uptake pattern of lactate, a known mediator of cell-cell interaction which was specifically altered in pericytes by the knockout of the key circadian regulator Bmal1. In an angiogenesis assay, the maturation of vessel-like structures was affected only when both endothelial cells and pericytes did not express Bmal1, indicating a compensation system. In a 3D tissue engineering scaffold, a synchronized clock supported a more structured organization of cells around the scaffold pores, and a maturation of vascular structures. Our results demonstrate that pericytes play a critical role in regulating the circadian rhythms in endothelial cells, and that silencing this system disproportionately affects their pro-angiogenic function. Particularly, in the context of tissue engineering and regenerative medicine, considering the effect of circadian rhythms may be critical for the development of mature vascular structures and to obtain the maximal reparative effect.
Collapse
Affiliation(s)
- Valeria Mastrullo
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
- Chronobiology Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, United Kingdom
| | - Daan R. van der Veen
- Chronobiology Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, United Kingdom
| | - Rolando S. Matos
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Jonathan D. Johnston
- Chronobiology Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - John H. McVey
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Eirini G. Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, United Kingdom
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London (UCL), London, United Kingdom
| | - Paola Campagnolo
- Cardiovascular Section, Department of Biochemical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
26
|
Gupta P, Miller A, Olayanju A, Madhuri TK, Velliou E. A Systematic Comparative Assessment of the Response of Ovarian Cancer Cells to the Chemotherapeutic Cisplatin in 3D Models of Various Structural and Biochemical Configurations-Does One Model Type Fit All? Cancers (Basel) 2022; 14:1274. [PMID: 35267582 PMCID: PMC8909317 DOI: 10.3390/cancers14051274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/18/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Epithelial Ovarian Cancer (EOC) is a silent, deadly and aggressive gynaecological disease with a relatively low survival rate. This has been attributed, to some extent, to EOC's high recurrence rate and resistance to currently available platinum-based chemotherapeutic treatment methods. Multiple groups have studied and reported the effect of chemotherapeutic agents on various EOC 3D in vitro models. However, there are very few studies wherein a direct comparative study has been carried out between the different in vitro 3D models of EOC and the effect of chemotherapy within them. Herein, we report, for the first time, a direct comprehensive systematic comparative study of three different 3D in vitro platforms, namely (i) spheroids, (ii) synthetic PeptiGels/hydrogels of various chemical configurations and (iii) polymeric scaffolds with coatings of various extracellular matrices (ECMs) on the cell growth and response to the chemotherapeutic (Cisplatin) for ovary-derived (A2780) and metastatic (SK-OV-3) EOC cell lines. We report that all three 3D models are able to support the growth of EOC, but for different time periods (varying from 7 days to 4 weeks). We have also reported that chemoresistance to Cisplatin, in vitro, observed especially for metastatic EOC cells, is platform-dependent, in terms of both the structural and biochemical composition of the model/platform. Our study highlights the importance of selecting an appropriate 3D platform for in vitro tumour model development. We have demonstrated that the selection of the best platform for producing in vitro tumour models depends on the cancer/cell type, the experimental time period and the application for which the model is intended.
Collapse
Affiliation(s)
- Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London W1W 7TY, UK;
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Surrey GU2 7XH, UK
| | - Aline Miller
- Manchester BIOGEL, 19F4, Mereside, Alderley Park, Alderley Edge, Chesire SK10 4TG, UK; (A.M.); (A.O.)
| | - Adedamola Olayanju
- Manchester BIOGEL, 19F4, Mereside, Alderley Park, Alderley Edge, Chesire SK10 4TG, UK; (A.M.); (A.O.)
| | - Thumuluru Kavitha Madhuri
- Department of Gynaecological Oncology Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK;
- Honorary Senior Lecturer in Cancer Research, School of Applied Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London W1W 7TY, UK;
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Surrey GU2 7XH, UK
| |
Collapse
|
27
|
Wishart G, Gupta P, Nisbet A, Schettino G, Velliou E. On the Evaluation of a Novel Hypoxic 3D Pancreatic Cancer Model as a Tool for Radiotherapy Treatment Screening. Cancers (Basel) 2021; 13:6080. [PMID: 34885188 PMCID: PMC8657010 DOI: 10.3390/cancers13236080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Tissue engineering is evolving to mimic intricate ecosystems of tumour microenvironments (TME) to more readily map realistic in vivo niches of cancerous tissues. Such advanced cancer tissue models enable more accurate preclinical assessment of treatment strategies. Pancreatic cancer is a dangerous disease with high treatment resistance that is directly associated with a highly complex TME. More specifically, the pancreatic cancer TME includes (i) complex structure and complex extracellular matrix (ECM) protein composition; (ii) diverse cell populations (e.g., stellate cells), cancer associated fibroblasts, endothelial cells, which interact with the cancer cells and promote resistance to treatment and metastasis; (iii) accumulation of high amounts of (ECM), which leads to the creation of a fibrotic/desmoplastic reaction around the tumour; and (iv) heterogeneous environmental gradients such as hypoxia, which result from vessel collapse and stiffness increase in the fibrotic/desmoplastic area of the TME. These unique hallmarks are not effectively recapitulated in traditional preclinical research despite radiotherapeutic resistance being largely connected to them. Herein, we investigate, for the first time, the impact of in vitro hypoxia (5% O2) on the radiotherapy treatment response of pancreatic cancer cells (PANC-1) in a novel polymer (polyurethane) based highly macroporous scaffold that was surface modified with proteins (fibronectin) for ECM mimicry. More specifically, PANC-1 cells were seeded in fibronectin coated macroporous scaffolds and were cultured for four weeks in in vitro normoxia (21% O2), followed by a two day exposure to either in vitro hypoxia (5% O2) or maintenance in in vitro normoxia. Thereafter, in situ post-radiation monitoring (one day, three days, seven days post-irradiation) of the 3D cell cultures took place via quantification of (i) live/dead and apoptotic profiles and (ii) ECM (collagen-I) and HIF-1a secretion by the cancer cells. Our results showed increased post-radiation viability, reduced apoptosis, and increased collagen-I and HIF-1a secretion in in vitro hypoxia compared to normoxic cultures, revealing hypoxia-induced radioprotection. Overall, this study employed a low cost, animal free model enabling (i) the possibility of long-term in vitro hypoxic 3D cell culture for pancreatic cancer, and (ii) in vitro hypoxia associated PDAC radio-protection development. Our novel platform for radiation treatment screening can be used for long-term in vitro post-treatment observations as well as for fractionated radiotherapy treatment.
Collapse
Affiliation(s)
- Gabrielle Wishart
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK;
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London (UCL), London WC1E 6BT, UK;
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK;
- National Physical Laboratory, Teddington TW11 0LW, UK
| | - Eirini Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.)
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| |
Collapse
|
28
|
In-vitro 3D modelling for charged particle therapy - Uncertainties and opportunities. Adv Drug Deliv Rev 2021; 179:114018. [PMID: 34688685 DOI: 10.1016/j.addr.2021.114018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 12/26/2022]
Abstract
Radiation therapy is a critical component of oncologic management, with more than half of all cancer patients requiring radiotherapy at some point during their disease course. Over the last decade, there has been increasing interest in charged particle therapy due to its advantageous physical and radiobiologic properties, with the therapeutic use of proton beam therapy (PBT) expanding worldwide. However, there remain large gaps in our knowledge of the radiobiologic mechanisms that underlie key aspects of PBT, such as variations in relative biologic effectiveness (RBE), radioresistance, DNA damage response and repair pathways, as well as immunologic effects. In addition, while the emerging technique of ultra-high dose rate or FLASH radiotherapy, with its potential to further reduce normal tissue toxicities, is an exciting development, in-depth study is needed into the postulated biochemical mechanisms that underpin the FLASH effect such as the oxygen depletion hypothesis as well as the relative contributions of immune responses and the tumor microenvironment. Further investigation is also required to ensure that the FLASH effect is not diminished or lost in PBT. Current methods to evaluate the biologic effects of charged particle therapy rely heavily on 2D cell culture systems and/or animal models. However, both of these methods have well-recognized limitations which limit translatability of findings from bench to bedside. The advent of novel three-dimensional in-vitro tumor models offers a more physiologically relevant and high throughput in-vitro system for the study of tumor development as well as novel therapeutic approaches such as PBT. Advances in 3D cell culture methods, together with knowledge of disease mechanism, biomarkers, and genomic data, can be used to design personalized 3D models that most closely recapitulate tumor microenvironmental factors promoting a particular disease phenotype, moving 3D models and PBT into the age of precision medicine.
Collapse
|
29
|
Lee KH, Kim TH. Recent Advances in Multicellular Tumor Spheroid Generation for Drug Screening. BIOSENSORS 2021; 11:445. [PMID: 34821661 PMCID: PMC8615712 DOI: 10.3390/bios11110445] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 05/12/2023]
Abstract
Multicellular tumor spheroids (MCTs) have been employed in biomedical fields owing to their advantage in designing a three-dimensional (3D) solid tumor model. For controlling multicellular cancer spheroids, mimicking the tumor extracellular matrix (ECM) microenvironment is important to understand cell-cell and cell-matrix interactions. In drug cytotoxicity assessments, MCTs provide better mimicry of conventional solid tumors that can precisely represent anticancer drug candidates' effects. To generate incubate multicellular spheroids, researchers have developed several 3D multicellular spheroid culture technologies to establish a research background and a platform using tumor modelingvia advanced materials science, and biosensing techniques for drug-screening. In application, drug screening was performed in both invasive and non-invasive manners, according to their impact on the spheroids. Here, we review the trend of 3D spheroid culture technology and culture platforms, and their combination with various biosensing techniques for drug screening in the biomedical field.
Collapse
Affiliation(s)
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Korea;
| |
Collapse
|
30
|
Zamorano M, Castillo RL, Beltran JF, Herrera L, Farias JA, Antileo C, Aguilar-Gallardo C, Pessoa A, Calle Y, Farias JG. Tackling Ischemic Reperfusion Injury With the Aid of Stem Cells and Tissue Engineering. Front Physiol 2021; 12:705256. [PMID: 34603075 PMCID: PMC8484708 DOI: 10.3389/fphys.2021.705256] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/11/2021] [Indexed: 01/14/2023] Open
Abstract
Ischemia is a severe condition in which blood supply, including oxygen (O), to organs and tissues is interrupted and reduced. This is usually due to a clog or blockage in the arteries that feed the affected organ. Reinstatement of blood flow is essential to salvage ischemic tissues, restoring O, and nutrient supply. However, reperfusion itself may lead to major adverse consequences. Ischemia-reperfusion injury is often prompted by the local and systemic inflammatory reaction, as well as oxidative stress, and contributes to organ and tissue damage. In addition, the duration and consecutive ischemia-reperfusion cycles are related to the severity of the damage and could lead to chronic wounds. Clinical pathophysiological conditions associated with reperfusion events, including stroke, myocardial infarction, wounds, lung, renal, liver, and intestinal damage or failure, are concomitant in due process with a disability, morbidity, and mortality. Consequently, preventive or palliative therapies for this injury are in demand. Tissue engineering offers a promising toolset to tackle ischemia-reperfusion injuries. It devises tissue-mimetics by using the following: (1) the unique therapeutic features of stem cells, i.e., self-renewal, differentiability, anti-inflammatory, and immunosuppressants effects; (2) growth factors to drive cell growth, and development; (3) functional biomaterials, to provide defined microarchitecture for cell-cell interactions; (4) bioprocess design tools to emulate the macroscopic environment that interacts with tissues. This strategy allows the production of cell therapeutics capable of addressing ischemia-reperfusion injury (IRI). In addition, it allows the development of physiological-tissue-mimetics to study this condition or to assess the effect of drugs. Thus, it provides a sound platform for a better understanding of the reperfusion condition. This review article presents a synopsis and discusses tissue engineering applications available to treat various types of ischemia-reperfusions, ultimately aiming to highlight possible therapies and to bring closer the gap between preclinical and clinical settings.
Collapse
Affiliation(s)
- Mauricio Zamorano
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | | | - Jorge F Beltran
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Lisandra Herrera
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Joaquín A Farias
- Facultad de Ingeniería y Ciencias, Universidad Adolfo Ibíñtez, Santiago, Chile
| | - Christian Antileo
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| | - Cristobal Aguilar-Gallardo
- Hematological Transplant and Cell Therapy Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Yolanda Calle
- Department of Life Sciences, Whitelands College, University of Roehampton, London, United Kingdom
| | - Jorge G Farias
- Department of Chemical Engineering, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
31
|
Prasad M, Kumar R, Buragohain L, Kumari A, Ghosh M. Organoid Technology: A Reliable Developmental Biology Tool for Organ-Specific Nanotoxicity Evaluation. Front Cell Dev Biol 2021; 9:696668. [PMID: 34631696 PMCID: PMC8495170 DOI: 10.3389/fcell.2021.696668] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
Engineered nanomaterials are bestowed with certain inherent physicochemical properties unlike their parent materials, rendering them suitable for the multifaceted needs of state-of-the-art biomedical, and pharmaceutical applications. The log-phase development of nano-science along with improved "bench to beside" conversion carries an enhanced probability of human exposure with numerous nanoparticles. Thus, toxicity assessment of these novel nanoscale materials holds a key to ensuring the safety aspects or else the global biome will certainly face a debacle. The toxicity may span from health hazards due to direct exposure to indirect means through food chain contamination or environmental pollution, even causing genotoxicity. Multiple ways of nanotoxicity evaluation include several in vitro and in vivo methods, with in vitro methods occupying the bulk of the "experimental space." The underlying reason may be multiple, but ethical constraints in in vivo animal experiments are a significant one. Two-dimensional (2D) monoculture is undoubtedly the most exploited in vitro method providing advantages in terms of cost-effectiveness, high throughput, and reproducibility. However, it often fails to mimic a tissue or organ which possesses a defined three-dimensional structure (3D) along with intercellular communication machinery. Instead, microtissues such as spheroids or organoids having a precise 3D architecture and proximate in vivo tissue-like behavior can provide a more realistic evaluation than 2D monocultures. Recent developments in microfluidics and bioreactor-based organoid synthesis have eased the difficulties to prosper nano-toxicological analysis in organoid models surpassing the obstacle of ethical issues. The present review will enlighten applications of organoids in nanotoxicological evaluation, their advantages, and prospects toward securing commonplace nano-interventions.
Collapse
Affiliation(s)
- Minakshi Prasad
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Rajesh Kumar
- Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, India
| | - Lukumoni Buragohain
- Department of Animal Biotechnology, College of Veterinary Science, Assam Agricultural University, Guwahati, India
| | | | - Mayukh Ghosh
- Department of Veterinary Physiology and Biochemistry, RGSC, Banaras Hindu University, Varanasi, India
| |
Collapse
|
32
|
Singh B, Abdelgawad ME, Ali Z, Bailey J, Budyn E, Civita P, Clift MJD, Connelly JT, Constant S, Hittinger M, Kandarova H, Kearns VR, Kiuru T, Kostrzewski T, Kress S, Durban VM, Lehr CM, McMillan H, Metz JK, Monteban V, Movia D, Neto C, Owen C, Paasonen L, Palmer KA, Pilkington GJ, Pilkington K, Prina-Mello A, Roper C, Sheard J, Smith S, Turner JE, Roy I, Tutty MA, Velliou E, Wilkinson JM. Towards More Predictive, Physiological and Animal-free In Vitro Models: Advances in Cell and Tissue Culture 2020 Conference Proceedings. Altern Lab Anim 2021; 49:93-110. [PMID: 34225465 DOI: 10.1177/02611929211025006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Experimental systems that faithfully replicate human physiology at cellular, tissue and organ level are crucial to the development of efficacious and safe therapies with high success rates and low cost. The development of such systems is challenging and requires skills, expertise and inputs from a diverse range of experts, such as biologists, physicists, engineers, clinicians and regulatory bodies. Kirkstall Limited, a biotechnology company based in York, UK, organised the annual conference, Advances in Cell and Tissue Culture (ACTC), which brought together people having a variety of expertise and interests, to present and discuss the latest developments in the field of cell and tissue culture and in vitro modelling. The conference has also been influential in engaging animal welfare organisations in the promotion of research, collaborative projects and funding opportunities. This report describes the proceedings of the latest ACTC conference, which was held virtually on 30th September and 1st October 2020, and included sessions on in vitro models in the following areas: advanced skin and respiratory models, neurological disease, cancer research, advanced models including 3-D, fluid flow and co-cultures, diabetes and other age-related disorders, and animal-free research. The roundtable session on the second day was very interactive and drew huge interest, with intriguing discussion taking place among all participants on the theme of replacement of animal models of disease.
Collapse
Affiliation(s)
| | - Mohamed Essameldin Abdelgawad
- Cellular, Molecular & Industrial Biotechnology and Cellular & Molecular Immunobiology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Zulfiqur Ali
- Healthcare Innovation Centre, School of Health and Life Sciences, Teesside University, Middlesbrough, UK
| | - Jarrod Bailey
- Center for Contemporary Sciences, Gaithersburg, MD, USA
| | - Elisa Budyn
- CNRS Laboratory of Mechanics and Technology, Ecole Normale Superieure Paris-Saclay, University Paris-Saclay, Gif-sur-Yvette, France
| | - Prospero Civita
- Brain Tumour Research Centre, Institute of Biological and Biomedical Sciences (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,School of Pharmacy and Pharmaceutical Sciences, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Martin J D Clift
- In Vitro Toxicology Group, Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - John T Connelly
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | - Helena Kandarova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Victoria Rosalind Kearns
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Tony Kiuru
- UPM-Kymmene Corporation, Helsinki, Finland
| | | | - Sebastian Kress
- Department of Biotechnology, Institute for Cell and Tissue Culture Technologies, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | - Claus-Michael Lehr
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), and Saarland University, Saarbrücken, Germany
| | - Hayley McMillan
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Julia Katharina Metz
- Pharmbiotec Research and Development GmbH, Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Dania Movia
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Catia Neto
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | | | | | - Kerri Anne Palmer
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | | | - Karen Pilkington
- School of Health and Social Care Professions, Faculty of Health and Science, University of Portsmouth, Portsmouth, UK
| | - Adriele Prina-Mello
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Clive Roper
- Roper Toxicology Consulting Limited, Edinburgh, UK
| | | | - Sheree Smith
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, UK
| | | | - Ipsita Roy
- Department of Materials Science & Engineering, Kroto Research Institute, University of Sheffield, Sheffield, UK
| | - Melissa Anne Tutty
- Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science-UCL, London, UK
| | | |
Collapse
|
33
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
34
|
Wishart G, Gupta P, Nisbet A, Velliou E, Schettino G. Novel Anticancer and Treatment Sensitizing Compounds against Pancreatic Cancer. Cancers (Basel) 2021; 13:2940. [PMID: 34208295 PMCID: PMC8231164 DOI: 10.3390/cancers13122940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
The isolation of chemical compounds from natural origins for medical application has played an important role in modern medicine with a range of novel treatments having emerged from various natural forms over the past decades. Natural compounds have been exploited for their antioxidant, antimicrobial and antitumor capabilities. Specifically, 60% of today's anticancer drugs originate from natural sources. Moreover, the combination of synthetic and natural treatments has shown applications for (i) reduced side effects, (ii) treatment sensitization and (iii) reduction in treatment resistance. This review aims to collate novel and natural compounds that are being explored for their preclinical anticancer, chemosensitizing and radiosensitizing effects on Pancreatic Ductal Adenocarcinoma (PDAC), which is a lethal disease with current treatments being inefficient and causing serve side effects. Two key points are highlighted by this work: (i) the availability of a range of natural compounds for potentially new therapeutic approaches for PDAC, (ii) potential synergetic impact of natural compounds with advanced chemo- and radio-therapeutic modalities for PDAC.
Collapse
Affiliation(s)
- Gabrielle Wishart
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, UK;
| | - Eirini Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK; (G.W.); (P.G.); (E.V.)
- Centre for 3D Models of Health and Disease, UCL-Division of Surgery and Interventional Science, Charles Bell House, 43-45 Foley Street, Fitzrovia, London W1W 7TY, UK
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford GU2 7XH, UK
- National Physical Laboratory, Teddington TW11 0LW, UK
| |
Collapse
|
35
|
Wishart G, Gupta P, Schettino G, Nisbet A, Velliou E. 3d tissue models as tools for radiotherapy screening for pancreatic cancer. Br J Radiol 2021; 94:20201397. [PMID: 33684308 PMCID: PMC8010544 DOI: 10.1259/bjr.20201397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
The efficiency of radiotherapy treatment regimes varies from tumour to tumour and from patient to patient but it is generally highly influenced by the tumour microenvironment (TME). The TME can be described as a heterogeneous composition of biological, biophysical, biomechanical and biochemical milieus that influence the tumour survival and its' response to treatment. Preclinical research faces challenges in the replication of these in vivo milieus for predictable treatment response studies. 2D cell culture is a traditional, simplistic and cost-effective approach to culture cells in vitro, however, the nature of the system fails to recapitulate important features of the TME such as structure, cell-cell and cell-matrix interactions. At the same time, the traditional use of animals (Xenografts) in cancer research allows realistic in vivo architecture, however foreign physiology, limited heterogeneity and reduced tumour mutation rates impairs relevance to humans. Furthermore, animal research is very time consuming and costly. Tissue engineering is advancing as a promising biomimetic approach, producing 3D models that capture structural, biophysical, biochemical and biomechanical features, therefore, facilitating more realistic treatment response studies for further clinical application. However, currently, the application of 3D models for radiation response studies is an understudied area of research, especially for pancreatic ductal adenocarcinoma (PDAC), a cancer with a notoriously complex microenvironment. At the same time, specific novel and/or more enhanced radiotherapy tumour-targeting techniques such as MRI-guided radiotherapy and proton therapy are emerging to more effectively target pancreatic cancer cells. However, these emerging technologies may have different biological effectiveness as compared to established photon-based radiotherapy. For example, for MRI-guided radiotherapy, the novel use of static magnetic fields (SMF) during radiation delivery is understudied and not fully understood. Thus, reliable biomimetic platforms to test new radiation delivery strategies are required to more accurately predict in vivo responses. Here, we aim to collate current 3D models for radiation response studies of PDAC, identifying the state of the art and outlines knowledge gaps. Overall, this review paper highlights the need for further research on the use of 3D models for pre-clinical radiotherapy screening including (i) 3D (re)-modeling of the PDAC hypoxic TME to allow for late effects of ionising radiation (ii) the screening of novel radiotherapy approaches and their combinations as well as (iii) a universally accepted 3D-model image quantification method for evaluating TME components in situ that would facilitate accurate post-treatment(s) quantitative comparisons.
Collapse
Affiliation(s)
| | - Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, UK
| | | | | | | |
Collapse
|
36
|
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: 25] [Impact Index Per Article: 6.3] [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
|
37
|
Ex vivo culture of intact human patient derived pancreatic tumour tissue. Sci Rep 2021; 11:1944. [PMID: 33479301 PMCID: PMC7820421 DOI: 10.1038/s41598-021-81299-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is attributed to the highly fibrotic stroma and complex multi-cellular microenvironment that is difficult to fully recapitulate in pre-clinical models. To fast-track translation of therapies and to inform personalised medicine, we aimed to develop a whole-tissue ex vivo explant model that maintains viability, 3D multicellular architecture, and microenvironmental cues of human pancreatic tumours. Patient-derived surgically-resected PDAC tissue was cut into 1-2 mm explants and cultured on gelatin sponges for 12 days. Immunohistochemistry revealed that human PDAC explants were viable for 12 days and maintained their original tumour, stromal and extracellular matrix architecture. As proof-of-principle, human PDAC explants were treated with Abraxane and we observed different levels of response between patients. PDAC explants were also transfected with polymeric nanoparticles + Cy5-siRNA and we observed abundant cytoplasmic distribution of Cy5-siRNA throughout the PDAC explants. Overall, our novel model retains the 3D architecture of human PDAC and has advantages over standard organoids: presence of functional multi-cellular stroma and fibrosis, and no tissue manipulation, digestion, or artificial propagation of organoids. This provides unprecedented opportunity to study PDAC biology including tumour-stromal interactions and rapidly assess therapeutic response to drive personalised treatment.
Collapse
|
38
|
Choi SR, Yang Y, Huang KY, Kong HJ, Flick MJ, Han B. Engineering of biomaterials for tumor modeling. MATERIALS TODAY. ADVANCES 2020; 8:100117. [PMID: 34541484 PMCID: PMC8448271 DOI: 10.1016/j.mtadv.2020.100117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Development of biomaterials mimicking tumor and its microenvironment has recently emerged for the use of drug discovery, precision medicine, and cancer biology. These biomimetic models have developed by reconstituting tumor and stroma cells within the 3D extracellular matrix. The models are recently extended to recapitulate the in vivo tumor microenvironment, including biological, chemical, and mechanical conditions tailored for specific cancer type and its microenvironment. In spite of the recent emergence of various innovative engineered tumor models, many of these models are still early stage to be adapted for cancer research. In this article, we review the current status of biomaterials engineering for tumor models considering three main aspects - cellular engineering, matrix engineering, and engineering for microenvironmental conditions. Considering cancer-specific variability in these aspects, our discussion is focused on pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC). In addition, we further discussed the current challenges and future opportunities to create reliable and relevant tumor models.
Collapse
Affiliation(s)
- Sae Rome Choi
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Yi Yang
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kai-Yu Huang
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyun Joon Kong
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|