1
|
Ferreira B, Barros AS, Leite-Pereira C, Viegas J, das Neves J, Nunes R, Sarmento B. Trends in 3D models of inflammatory bowel disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167042. [PMID: 38296115 DOI: 10.1016/j.bbadis.2024.167042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/11/2023] [Accepted: 01/25/2024] [Indexed: 02/20/2024]
Abstract
Inflammatory bowel disease (IBD) encompasses a set of chronic inflammatory conditions, namely Crohn's disease and ulcerative colitis. Despite all advances in the management of IBD, a definitive cure is not available, largely due to a lack of a holistic understanding of its etiology and pathophysiology. Several in vitro, in vivo, and ex vivo models have been developed over the past few decades in order to abbreviate remaining gaps. The establishment of reliable and predictable in vitro intestinal inflammation models may indeed provide valuable tools to expedite and validate the development of therapies for IBD. Three-dimensional (3D) models provide a more accurate representation of the different layers of the intestine, contributing to a stronger impact on drug screening and research on intestinal inflammation, and bridging the gap between in vitro and in vivo research. This work provides a critical overview on the state-of-the-art on existing 3D models of intestinal inflammation and discusses the remaining challenges, providing insights on possible pathways towards achieving IBD mimetic models. We also address some of the main challenges faced by implementing cell culture models in IBD research while bearing in mind clinical translational aspects.
Collapse
Affiliation(s)
- Bárbara Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Andreia S Barros
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Catarina Leite-Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Rute Nunes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS-CESPU - Instituto Universitário de Ciências da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
2
|
Larsson AT, Bhatia H, Calizo A, Pollard K, Zhang X, Conniff E, Tibbitts JF, Rono E, Cummins K, Osum SH, Williams KB, Crampton AL, Jubenville T, Schefer D, Yang K, Lyu Y, Pino JC, Bade J, Gross JM, Lisok A, Dehner CA, Chrisinger JSA, He K, Gosline SJC, Pratilas CA, Largaespada DA, Wood DK, Hirbe AC. Ex vivo to in vivo model of malignant peripheral nerve sheath tumors for precision oncology. Neuro Oncol 2023; 25:2044-2057. [PMID: 37246765 PMCID: PMC10628938 DOI: 10.1093/neuonc/noad097] [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: 05/16/2022] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft tissue sarcomas that often develop in patients with neurofibromatosis type 1 (NF1). To address the critical need for novel therapeutics in MPNST, we aimed to establish an ex vivo 3D platform that accurately captured the genomic diversity of MPNST and could be utilized in a medium-throughput manner for drug screening studies to be validated in vivo using patient-derived xenografts (PDX). METHODS Genomic analysis was performed on all PDX-tumor pairs. Selected PDX were harvested for assembly into 3D microtissues. Based on prior work in our labs, we evaluated drugs (trabectedin, olaparib, and mirdametinib) ex vivo and in vivo. For 3D microtissue studies, cell viability was the endpoint as assessed by Zeiss Axio Observer. For PDX drug studies, tumor volume was measured twice weekly. Bulk RNA sequencing was performed to identify pathways enriched in cells. RESULTS We developed 13 NF1-associated MPNST-PDX and identified mutations or structural abnormalities in NF1 (100%), SUZ12 (85%), EED (15%), TP53 (15%), CDKN2A (85%), and chromosome 8 gain (77%). We successfully assembled PDX into 3D microtissues, categorized as robust (>90% viability at 48 h), good (>50%), or unusable (<50%). We evaluated drug response to "robust" or "good" microtissues, namely MN-2, JH-2-002, JH-2-079-c, and WU-225. Drug response ex vivo predicted drug response in vivo, and enhanced drug effects were observed in select models. CONCLUSIONS These data support the successful establishment of a novel 3D platform for drug discovery and MPNST biology exploration in a system representative of the human condition.
Collapse
Affiliation(s)
- Alex T Larsson
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Himanshi Bhatia
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ana Calizo
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiaochun Zhang
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Justin F Tibbitts
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara H Osum
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kyle B Williams
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexandra L Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tyler Jubenville
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel Schefer
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kuangying Yang
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yang Lyu
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | - James C Pino
- Pacific Northwest National Laboratory, Seattle, Washington, USA
| | - Jessica Bade
- Pacific Northwest National Laboratory, Seattle, Washington, USA
| | - John M Gross
- Department of Pathology, Division of Surgical Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Alla Lisok
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carina A Dehner
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri, USA
| | - John S A Chrisinger
- Department of Pathology and Immunology, Washington University in St. Louis, Missouri, USA
| | - Kevin He
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David A Largaespada
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela C Hirbe
- Division of Oncology, Department of Internal Medicine, Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Davy M, Genest L, Legrand C, Pelouin O, Froget G, Castagné V, Rupp T. Evaluation of Temozolomide and Fingolimod Treatments in Glioblastoma Preclinical Models. Cancers (Basel) 2023; 15:4478. [PMID: 37760448 PMCID: PMC10527257 DOI: 10.3390/cancers15184478] [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: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastomas are malignant brain tumors which remain lethal due to their aggressive and invasive nature. The standard treatment combines surgical resection, radiotherapy, and chemotherapy using Temozolomide, albeit with a minor impact on patient prognosis (15 months median survival). New therapies evaluated in preclinical translational models are therefore still required to improve patient survival and quality of life. In this preclinical study, we evaluated the effect of Temozolomide in different models of glioblastoma. We also aimed to investigate the efficacy of Fingolimod, an immunomodulatory drug for multiple sclerosis also described as an inhibitor of the sphingosine-1-phosphate (S1P)/S1P receptor axis. The effects of Fingolimod and Temozolomide were analyzed with in vitro 2D and 3D cellular assay and in vivo models using mouse and human glioblastoma cells implanted in immunocompetent or immunodeficient mice, respectively. We demonstrated both in in vitro and in vivo models that Temozolomide has a varied effect depending on the tumor type (i.e., U87MG, U118MG, U138MG, and GL261), demonstrating sensitivity, acquired resistance, and purely resistant tumor phenotypes, as observed in patients. Conversely, Fingolimod only reduced in vitro 2D tumor cell growth and increased cytotoxicity. Indeed, Fingolimod had little or no effect on 3D spheroid cytotoxicity and was devoid of effect on in vivo tumor progression in Temozolomide-sensitive models. These results suggest that the efficacy of Fingolimod is dependent on the glioblastoma tumor microenvironment. Globally, our data suggest that the response to Temozolomide varies depending on the cancer model, consistent with its clinical activity, whereas the potential activity of Fingolimod may merit further evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tristan Rupp
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France
| |
Collapse
|
4
|
Modi U, Kedaria D, Vasita R. Differential Migration and Proliferation Potential of the Hydrogel Aided 3D Tumoroid. Macromol Biosci 2022; 22:e2200196. [PMID: 35997309 DOI: 10.1002/mabi.202200196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/04/2022] [Indexed: 12/25/2022]
Abstract
For substantial in vitro cancer biology research, the 3D cell culture method has now been regarded as more suitable model expected to be recapitulating maximum in vivo tumor mass relevance. Despite of available techniques to develop in vitro 3D models, a system availing a physiologically relevant in vitro 3D model of primary lung adenocarcinoma with extracellular matrix (ECM) mimicry and similar tumorigenic properties still remains a quest. Thus, in the present study, chemically modified Dextran-Chitosan (MDC) hydrogel has been developed as a 3D tumoroid aiding scaffold. The 3D A549 tumoroids aided by the MDC scaffold have physiologically relevant proliferation, migration, invasive potential, and Gefitinib [targeting epidermal growth factor receptor (EGFR)] efficacy as compared to the 2D cultured cells. The surface topography and wettability of hydrogel availed in vivo micro tumor mass mimicking Lung adenocarcinoma 3D in vitro model. Thus, opening an innovative avenue for elucidating the disease mechanism and drug efficacy on relevant 3D cancer models in vitro.
Collapse
Affiliation(s)
- Unnati Modi
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Dhaval Kedaria
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| | - Rajesh Vasita
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, 382030, India
| |
Collapse
|
5
|
Valančiūtė A, Mathieson L, O’Connor RA, Scott JI, Vendrell M, Dorward DA, Akram AR, Dhaliwal K. Phototherapeutic Induction of Immunogenic Cell Death and CD8+ T Cell-Granzyme B Mediated Cytolysis in Human Lung Cancer Cells and Organoids. Cancers (Basel) 2022; 14:4119. [PMID: 36077656 PMCID: PMC9454585 DOI: 10.3390/cancers14174119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Augmenting T cell mediated tumor killing via immunogenic cancer cell death (ICD) is the cornerstone of emerging immunotherapeutic approaches. We investigated the potential of methylene blue photodynamic therapy (MB-PDT) to induce ICD in human lung cancer. Non-Small Cell Lung Cancer (NSCLC) cell lines and primary human lung cancer organoids were evaluated in co-culture killing assays with MB-PDT and light emitting diodes (LEDs). ICD was characterised using immunoblotting, immunofluorescence, flow cytometry and confocal microscopy. Phototherapy with MB treatment and low energy LEDs decreased the proliferation of NSCLC cell lines inducing early necrosis associated with reduced expression of the anti-apoptotic protein, Bcl2 and increased expression of ICD markers, calreticulin (CRT), intercellular cell-adhesion molecule-1 (ICAM-1) and major histocompatibility complex I (MHC-I) in NSCLC cells. MB-PDT also potentiated CD8+ T cell-mediated cytolysis of lung cancer via granzyme B in lung cancer cells and primary human lung cancer organoids.
Collapse
Affiliation(s)
- Asta Valančiūtė
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Layla Mathieson
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard A. O’Connor
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jamie I. Scott
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Marc Vendrell
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - David A. Dorward
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Ahsan R. Akram
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kevin Dhaliwal
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
6
|
Moya-Garcia CR, Okuyama H, Sadeghi N, Li J, Tabrizian M, Li-Jessen NYK. In vitro models for head and neck cancer: Current status and future perspective. Front Oncol 2022; 12:960340. [PMID: 35992863 PMCID: PMC9381731 DOI: 10.3389/fonc.2022.960340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The 5-year overall survival rate remains approximately 50% for head and neck (H&N) cancer patients, even though new cancer drugs have been approved for clinical use since 2016. Cancer drug studies are now moving toward the use of three-dimensional culture models for better emulating the unique tumor microenvironment (TME) and better predicting in vivo response to cancer treatments. Distinctive TME features, such as tumor geometry, heterogenous cellularity, and hypoxic cues, notably affect tissue aggressiveness and drug resistance. However, these features have not been fully incorporated into in vitro H&N cancer models. This review paper aims to provide a scholarly assessment of the designs, contributions, and limitations of in vitro models in H&N cancer drug research. We first review the TME features of H&N cancer that are most relevant to in vitro drug evaluation. We then evaluate a selection of advanced culture models, namely, spheroids, organotypic models, and microfluidic chips, in their applications for H&N cancer drug research. Lastly, we propose future opportunities of in vitro H&N cancer research in the prospects of high-throughput drug screening and patient-specific drug evaluation.
Collapse
Affiliation(s)
| | - Hideaki Okuyama
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
- Department of Otolaryngology – Head & Neck Surgery, Kyoto University, Kyoto, Japan
| | - Nader Sadeghi
- Department of Otolaryngology – Head and Neck Surgery, McGill University, Montreal, QC, Canada
- Research Institute of McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Jianyu Li
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- *Correspondence: Maryam Tabrizian, ; Nicole Y. K. Li-Jessen,
| | - Nicole Y. K. Li-Jessen
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
- Department of Otolaryngology – Head and Neck Surgery, McGill University, Montreal, QC, Canada
- Research Institute of McGill University Health Center, McGill University, Montreal, QC, Canada
- *Correspondence: Maryam Tabrizian, ; Nicole Y. K. Li-Jessen,
| |
Collapse
|
7
|
Rupp T, Debasly S, Genest L, Froget G, Castagné V. Therapeutic Potential of Fingolimod and Dimethyl Fumarate in Non-Small Cell Lung Cancer Preclinical Models. Int J Mol Sci 2022; 23:ijms23158192. [PMID: 35897763 PMCID: PMC9330228 DOI: 10.3390/ijms23158192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 01/27/2023] Open
Abstract
New therapies are required for patients with non-small cell lung cancer (NSCLC) for which the current standards of care poorly affect the patient prognosis of this aggressive cancer subtype. In this preclinical study, we aim to investigate the efficacy of Fingolimod, a described inhibitor of sphingosine-1-phosphate (S1P)/S1P receptors axis, and Dimethyl Fumarate (DMF), a methyl ester of fumaric acid, both already approved as immunomodulators in auto-immune diseases with additional expected anti-cancer effects. The impact of both drugs was analyzed with in vitro cell survival analysis and in vivo graft models using mouse and human NSCLC cells implanted in immunocompetent or immunodeficient mice, respectively. We demonstrated that Fingolimod and DMF repressed tumor progression without apparent adverse effects in vivo in three preclinical mouse NSCLC models. In vitro, Fingolimod did not affect either the tumor proliferation or the cytotoxicity, although DMF reduced tumor cell proliferation. These results suggest that Fingolimod and DMF affected tumor progression through different cellular mechanisms within the tumor microenvironment. Fingolimod and DMF might uncover potential therapeutic opportunities in NSCLC.
Collapse
Affiliation(s)
- Tristan Rupp
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
- Correspondence: or ; Tel.: +33-(0)2-43-69-36-07
| | - Solène Debasly
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
- CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Université de Reims-Champagne-Ardenne, Campus Moulin de la Housse, 51687 Reims, France
| | - Laurie Genest
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| | - Guillaume Froget
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| | - Vincent Castagné
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| |
Collapse
|
8
|
Powell RT, Moussalli MJ, Guo L, Bae G, Singh P, Stephan C, Shureiqi I, Davies PJ. deepOrganoid: A brightfield cell viability model for screening matrix-embedded organoids. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:175-184. [PMID: 35314378 DOI: 10.1016/j.slasd.2022.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
High-throughput viability screens are commonly used in the identification and development of chemotherapeutic drugs. These systems rely on the fidelity of the cellular model systems to recapitulate the drug response that occurs in vivo. In recent years, there has been an expansion in the utilization of patient-derived materials as well as advanced cell culture techniques, such as multi-cellular tumor organoids, to further enhance the translational relevance of cellular model systems. Simple quantitative analysis remains a challenge, primarily due to the difficulties of robust image segmentation in heterogenous 3D cultures. However, explicit segmentation is not required with the advancement of deep learning, and it can be used for both continuous (regression) or categorical classification problems. Deep learning approaches are additionally benefited by being fully data-driven and highly automatable, thus they can be established and run with minimal to no user-defined parameters. In this article, we describe the development and implementation of a regressive deep learning model trained on brightfield images of patient-derived organoids and use the terminal viability readout (CellTiter-Glo) as training labels. Ultimately, this has led to the generation of a non-invasive and label-free tool to evaluate changes in organoid viability.
Collapse
Affiliation(s)
- Reid T Powell
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States.
| | - Micheline J Moussalli
- Department of Gastrointestinal Medical Oncology, UT MDACC, Houston, TX, United States
| | - Lei Guo
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Goeun Bae
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Pankaj Singh
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Clifford Stephan
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, UT MDACC, Houston, TX, United States
| | - Peter J Davies
- Center for Translational Cancer Research, Texas A&M University, 2121 W. Holcombe Blvd. Rm 911, Houston, TX 77030, United States
| |
Collapse
|
9
|
Drug-Induced Liver Injury: Clinical Evidence of N-Acetyl Cysteine Protective Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3320325. [PMID: 34912495 PMCID: PMC8668310 DOI: 10.1155/2021/3320325] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022]
Abstract
Oxidative stress is a key pathological feature implicated in both acute and chronic liver diseases, including drug-induced liver injury (DILI). The latter describes hepatic injury arising as a direct toxic effect of administered drugs or their metabolites. Although still underreported, DILI remains a significant cause of liver failure, especially in developed nations. Currently, it is understood that mitochondrial-generated oxidative stress and abnormalities in phase I/II metabolism, leading to glutathione (GSH) suppression, drive the onset of DILI. N-Acetyl cysteine (NAC) has attracted a lot of interest as a therapeutic agent against DILI because of its strong antioxidant properties, especially in relation to enhancing endogenous GSH content to counteract oxidative stress. Thus, in addition to updating information on the pathophysiological mechanisms implicated in oxidative-induced hepatic injury, the current review critically discusses clinical evidence on the protective effects of NAC against DILI, including the reduction of patient mortality. Besides injury caused by paracetamol, NAC can also improve liver function in relation to other forms of liver injury such as those induced by excessive alcohol intake. The implicated therapeutic mechanisms of NAC extend from enhancing hepatic GSH levels to reducing biomarkers of paracetamol toxicity such as keratin-18 and circulating caspase-cleaved cytokeratin-18. However, there is still lack of evidence confirming the benefits of using NAC in combination with other therapies in patients with DILI.
Collapse
|
10
|
Almadori G, Coli A, De Corso E, Mele DA, Settimi S, Di Cintio G, Brigato F, Scannone D, Carey TE, Paludetti G, Lauriola L, Ranelletti FO. Nuclear HER3 expression improves the prognostic stratification of patients with HER1 positive advanced laryngeal squamous cell carcinoma. J Transl Med 2021; 19:408. [PMID: 34579737 PMCID: PMC8477517 DOI: 10.1186/s12967-021-03081-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/14/2021] [Indexed: 11/24/2022] Open
Abstract
Background Compared to the other members of human epidermal growth factor family receptors (HER), the role of HER3 has not been well defined in laryngeal cancer. The predictive and prognostic role of HER3 has been the focus of clinical attention but the research findings are contradictory, especially in laryngeal squamous cell carcinoma (LSCC). The variable localization of HER3 within cancer cells and the role of HER3 in primary and acquired resistance to HER1-targeted therapies remain unclear. Methods We performed a retrospective analysis of two cohorts of 66 homogeneous consecutive untreated primary advanced LSCC patients, in which co-expression of HER1, HER2 and HER3 receptors was investigated by semi-quantitative immunohistochemistry. The association of their pattern of expression with survival was evaluated by Kaplan–Meier and Cox’s proportional hazard analyses. Multivariable Cox proportional hazards models were developed to predict median 2- and 3-year RFS and 2.5- and 5-year OS. The Akaike information criterion technique and backwards stepwise procedure were used for model selections. The performance of the final Cox models was assessed with respect to calibration and discrimination. Results Immunohistochemical labeling for HER1 and HER2 was localized both in the cell membrane and in the cytoplasm, while HER3 labeling was observed both in the cell cytoplasm and in the nucleus. HER3 expression was inversely correlated with HER1 positivity. The expression patterns of HERs were associated with tumor differentiation. In both cohorts of patients, HER1 expression was associated with reduced relapse-free (RFS) and overall survival (OS). In HER1 positive tumors, the co-expression with nuclear HER3 was associated with better RFS and OS, compared with HER3 negative tumors or tumors expressing HER3 at cytoplasmic level. HER3 expressing tumors had a higher Geminin/MCM7 ratio than HER3 negative ones, regardless of HER1 co-expression. Multivariable analyses identified age at diagnosis, tumor site, HER1, HER3 and age at diagnosis, tumor stage, HER1, HER3, as covariates significantly associated with RFS and OS, respectively. Bootstrapping verified the good fitness of these models for predicting survivals and the optimism-corrected C-indices were 0.76 and 0.77 for RFS and OS, respectively. Conclusions Nuclear HER3 expression was strongly associated with favourable prognosis and allows to improve the prognostic stratification of patients with HER1 positive advanced LSCC carcinoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03081-0.
Collapse
Affiliation(s)
- Giovanni Almadori
- Unit of Head and Neck Oncology, "A. Gemelli" University Hospital Foundation IRCCS-Catholic University of the Sacred Heart, Largo A. Gemelli 8, 00168, Rome, Italy. .,Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy. .,Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Antonella Coli
- Università Cattolica del Sacro Cuore, Roma, Italy.,Unit of Anatomic Pathology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Eugenio De Corso
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Dario Antonio Mele
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Stefano Settimi
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Giovanni Di Cintio
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Francesca Brigato
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Domenico Scannone
- Unit of Anatomic Pathology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | - Thomas E Carey
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Gaetano Paludetti
- Unit of Otorhinolaryngology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy.,Università Cattolica del Sacro Cuore, Roma, Italy
| | - Libero Lauriola
- Università Cattolica del Sacro Cuore, Roma, Italy.,Unit of Anatomic Pathology, "A. Gemelli" University Hospital Foundation IRCCS, Roma, Italy
| | | |
Collapse
|
11
|
Franchi-Mendes T, Eduardo R, Domenici G, Brito C. 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers (Basel) 2021; 13:4610. [PMID: 34572836 PMCID: PMC8468887 DOI: 10.3390/cancers13184610] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
The tumour microenvironment plays a critical role in tumour progression and drug resistance processes. Non-malignant cell players, such as fibroblasts, endothelial cells, immune cells and others, interact with each other and with the tumour cells, shaping the disease. Though the role of each cell type and cell communication mechanisms have been progressively studied, the complexity of this cellular network and its role in disease mechanism and therapeutic response are still being unveiled. Animal models have been mainly used, as they can represent systemic interactions and conditions, though they face recognized limitations in translational potential due to interspecies differences. In vitro 3D cancer models can surpass these limitations, by incorporating human cells, including patient-derived ones, and allowing a range of experimental designs with precise control of each tumour microenvironment element. We summarize the role of each tumour microenvironment component and review studies proposing 3D co-culture strategies of tumour cells and non-malignant cell components. Moreover, we discuss the potential of these modelling approaches to uncover potential therapeutic targets in the tumour microenvironment and assess therapeutic efficacy, current bottlenecks and perspectives.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Rodrigo Eduardo
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Giacomo Domenici
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
12
|
Hirashita Y, Tsukamoto Y, Kudo Y, Kakisako D, Kurogi S, Hijiya N, Nakada C, Uchida T, Hirashita T, Hiratsuka T, Akagi T, Ueda Y, Shiroshita H, Etoh T, Mizukami K, Honda K, Okimoto T, Kodama M, Inomata M, Moriyama M, Murakami K. Early response in phosphorylation of ribosomal protein S6 is associated with sensitivity to trametinib in colorectal cancer cells. J Transl Med 2021; 101:1036-1047. [PMID: 33911189 DOI: 10.1038/s41374-021-00590-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/09/2022] Open
Abstract
Mutations in RAS or BRAF are associated with poor prognosis and resistance to epidermal growth factor receptor (EGFR)-targeted therapy in colorectal cancer (CRC). Despite their common ability to activate downstream genes such as MEK and ERK, the therapeutic benefit of MEK inhibitors for patients with RAS/BRAF mutant CRC is limited, highlighting the need for biomarkers to predict the efficacy of MEK inhibition. Previously, we reported that a change in phosphorylation of ribosomal protein S6 (pS6) after MEK inhibition was significantly associated with sensitivity to MEK inhibition in gastric cancer cells. Here, we investigated the value of the response in pS6 for predicting the efficacy of trametinib, a MEK inhibitor, in patients with RAS/BRAF mutant CRC using patient-derived CRC organoids. We found that a subset of CRC cell lines and organoids were sensitive to trametinib. The change in phosphorylated ERK, a downstream molecule of the RAS/RAF/MEK pathway, was not significantly associated with trametinib sensitivity. On the other hand, only those with sensitivity showed a reduction of pS6 levels in response to trametinib. The change in pS6 after trametinib treatment was detectable by Western blotting, immunohistochemistry or immunocytochemistry. We also demonstrated an impact of MEK inhibition on pS6 in vivo using a xenograft model. Our data suggest that, in combination with patient-derived organoids, immunostaining-based detection of pS6 could be useful for prediction of trametinib sensitivity.
Collapse
Affiliation(s)
- Yuka Hirashita
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan.
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan.
| | - Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan.
| | - Yoko Kudo
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Daisuke Kakisako
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Shusaku Kurogi
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Naoki Hijiya
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Chisato Nakada
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Tomohisa Uchida
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Teijiro Hirashita
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Takahiro Hiratsuka
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Tomonori Akagi
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshitake Ueda
- Department of Comprehensive Surgery for Community Medicine, Oita University, Oita, Japan
| | - Hidefumi Shiroshita
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Tsuyoshi Etoh
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Koichi Honda
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Tadayoshi Okimoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masaaki Kodama
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masafumi Inomata
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Masatsugu Moriyama
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
13
|
Zhuang Y, Grainger JM, Vedell PT, Yu J, Moyer AM, Gao H, Fan XY, Qin S, Liu D, Kalari KR, Goetz MP, Boughey JC, Weinshilboum RM, Wang L. Establishment and characterization of immortalized human breast cancer cell lines from breast cancer patient-derived xenografts (PDX). NPJ Breast Cancer 2021; 7:79. [PMID: 34145270 PMCID: PMC8213738 DOI: 10.1038/s41523-021-00285-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
The application of patient-derived xenografts (PDX) in drug screening and testing is a costly and time-consuming endeavor. While cell lines permit extensive mechanistic studies, many human breast cancer cell lines lack patient characteristics and clinical treatment information. Establishing cell lines that retain patient's genetic and drug response information would enable greater drug screening and mechanistic studies. Therefore, we utilized breast cancer PDX from the Mayo Breast Cancer Genome Guided Therapy Study (BEAUTY) to establish two immortalized, genomically unique breast cancer cell lines. Through extensive genetic and therapeutic testing, the cell lines were found to retain the same clinical subtype, major somatic alterations, and drug response phenotypes as their corresponding PDX and patient tumor. Our findings demonstrate PDX can be utilized to develop immortalized breast cancer cell lines and provide a valuable tool for understanding the molecular mechanism of drug resistance and exploring novel treatment strategies.
Collapse
Affiliation(s)
- Yongxian Zhuang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jordan M Grainger
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Peter T Vedell
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jia Yu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ann M Moyer
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Huanyao Gao
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Xiao-Yang Fan
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Sisi Qin
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Matthew P Goetz
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Concentration-Dependent Pro- and Antitumor Activities of Quercetin in Human Melanoma Spheroids: Comparative Analysis of 2D and 3D Cell Culture Models. Molecules 2021; 26:molecules26030717. [PMID: 33573155 PMCID: PMC7866537 DOI: 10.3390/molecules26030717] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 01/06/2023] Open
Abstract
Quercetin, a dietary flavonoid found in fruits and vegetables, has been described as a substance with many anti-cancer properties in a variety of preclinical investigations. In the present study, we demonstrate that 2D and 3D melanoma models exhibit not only different sensitivities to quercetin, but also opposite, cancer-promoting effects when metastatic melanoma spheroids are treated with quercetin. Higher concentrations of quercetin reduce melanoma growth in three tested cell lines, whereas low concentrations induce the opposite effect in metastatic melanoma spheroids but not in the non-metastatic cell line. High (>12.5 µM) or low (<6.3 µM) quercetin concentrations decrease or enhance cell viability, spheroid size, and cell proliferation, respectively. Additionally, melanoma cells cultivated in 2D already show significant caspase 3 activity at very low concentrations (>0.4 µM), whereas in 3D spheroids apoptotic cells, caspase 3 activity can only be detected in concentrations ≥12.5 µM. Further, we show that the tumor promoting or repressing effect in the 3D metastatic melanoma spheroids are likely to be elicited by a precisely controlled regulation of Nrf2/ARE-mediated cytoprotective genes, as well as ERK and NF-κB phosphorylation. According to the results obtained here, further studies are needed to better characterize the mechanisms of action underlying the pro- and anti-carcinogenic effects of quercetin on human melanomas.
Collapse
|
15
|
Ntamo Y, Samodien E, Burger J, Muller N, Muller CJF, Chellan N. In vitro Characterization of Insulin-Producing β-Cell Spheroids. Front Cell Dev Biol 2021; 8:623889. [PMID: 33585464 PMCID: PMC7876261 DOI: 10.3389/fcell.2020.623889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022] Open
Abstract
Over the years, immortalized rodent β-cell lines such as RIN, HIT, MIN, βTC, and INS-1 have been used to investigate pancreatic β-cell physiology using conventional two-dimensional (2D) culture techniques. However, physical and physiological limitations inherent to 2D cell culture necessitates confirmatory follow up studies using sentient animals. Three-dimensional (3D) culture models are gaining popularity for their recapitulation of key features of in vivo organ physiology, and thus could pose as potential surrogates for animal experiments. In this study, we aimed to develop and characterize a rat insulinoma INS-1 3D spheroid model to compare with 2D monolayers of the same cell line. Ultrastructural verification was done by transmission electron microscopy and toluidine blue staining, which showed that both 2D monolayers and 3D spheroids contained highly granulated cells with ultrastructural features synonymous with mature pancreatic β-cells, with increased prominence of these features observed in 3D spheroids. Viability, as assessed by cellular ATP quantification, size profiling and glucose utilization, showed that our spheroids remained viable for the experimental period of 30 days, compared to the limiting 5-day passage period of INS-1 monolayers. In fact, increasing ATP content together with spheroid size was observed over time, without adverse changes in glucose utilization. Additionally, β-cell function, assessed by determining insulin and amylin secretion, showed that the 3D spheroids retained glucose sensing and insulin secretory capability, that was more acute when compared to 2D monolayer cultures. Thus, we were able to successfully demonstrate that our in vitro INS-1 β-cell 3D spheroid model exhibits in vivo tissue-like structural features with extended viability and lifespan. This offers enhanced predictive capacity of the model in the study of metabolic disease, β-cell pathophysiology and the potential treatment thereof.
Collapse
Affiliation(s)
- Yonela Ntamo
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, South Africa
| | - Ebrahim Samodien
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa
| | - Joleen Burger
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nolan Muller
- National Health Laboratory Service, Anatomical Pathology, Tygerberg Hospital, Cape Town, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nireshni Chellan
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
16
|
Salman MI, Emran MA, Al-Shammari AM. Spheroid-formation 3D engineering model assay for in vitro assessment and expansion of cancer cells. AIP CONFERENCE PROCEEDINGS 2021. [DOI: 10.1063/5.0065362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
17
|
Bhaumik S, Boyer J, Banerjee C, Clark S, Sebastiao N, Vela E, Towne P. Fluorescent multiplexing of 3D spheroids: Analysis of biomarkers using automated immunohistochemistry staining platform and multispectral imaging. J Cell Biochem 2020; 121:4974-4990. [PMID: 32692912 PMCID: PMC7689845 DOI: 10.1002/jcb.29827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/24/2020] [Accepted: 06/23/2020] [Indexed: 11/25/2022]
Abstract
In preclinical cancer studies, three-dimensional (3D) cell spheroids and aggregates are preferred over monolayer cell cultures due to their architectural and functional similarity to solid tumors. We performed a proof-of-concept study to generate physiologically relevant and predictive preclinical models using non-small cell lung adenocarcinoma, and colon and colorectal adenocarcinoma cell line-derived 3D spheroids and aggregates. Distinct panels were designed to determine the expression profiles of frequently studied biomarkers of the two cancer subtypes. The lung adenocarcinoma panel included ALK, EGFR, TTF-1, and CK7 biomarkers, and the colon and colorectal adenocarcinoma panel included BRAF V600E, MSH2, MSH6, and CK20. Recent advances in immunofluorescence (IF) multiplexing and imaging technology enable simultaneous detection and quantification of multiple biomarkers on a single slide. In this study, we performed IF staining of multiple biomarkers per section on formalin-fixed paraffin-embedded 3D spheroids and aggregates. We optimized protocol parameters for automated IF and demonstrated staining concordance with automated chromogenic immunohistochemistry performed with validated protocols. Next, post-acquisition spectral unmixing of the captured fluorescent signals were utilized to delineate four differently stained biomarkers within a single multiplex IF image, followed by automated quantification of the expressed markers. This workflow has the potential to be adapted to preclinical high-throughput screening and drug efficacy studies utilizing 3D spheroids from cancer cell lines and patient-derived organoids. The process allows for cost, time, and resource savings through concurrent staining of several biomarkers on a single slide, the ability to study the interactions of multiple expressed proteins within a single region of interest, and enable quantitative assessment of biomarkers in cancer cells.
Collapse
|
18
|
Theard PL, Sheffels E, Sealover NE, Linke AJ, Pratico DJ, Kortum RL. Marked synergy by vertical inhibition of EGFR signaling in NSCLC spheroids shows SOS1 is a therapeutic target in EGFR-mutated cancer. eLife 2020; 9:58204. [PMID: 32897190 PMCID: PMC7478890 DOI: 10.7554/elife.58204] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/14/2020] [Indexed: 01/13/2023] Open
Abstract
Drug treatment of 3D cancer spheroids more accurately reflects in vivo therapeutic responses compared to adherent culture studies. In EGFR-mutated lung adenocarcinoma, EGFR-TKIs show enhanced efficacy in spheroid cultures. Simultaneous inhibition of multiple parallel RTKs further enhances EGFR-TKI effectiveness. We show that the common RTK signaling intermediate SOS1 was required for 3D spheroid growth of EGFR-mutated NSCLC cells. Using two distinct measures of pharmacologic synergy, we demonstrated that SOS1 inhibition strongly synergized with EGFR-TKI treatment only in 3D spheroid cultures. Combined EGFR- and SOS1-inhibition markedly inhibited Raf/MEK/ERK and PI3K/AKT signaling. Finally, broad assessment of the pharmacologic landscape of drug-drug interactions downstream of mutated EGFR revealed synergy when combining an EGFR-TKI with inhibitors of proximal signaling intermediates SOS1 and SHP2, but not inhibitors of downstream RAS effector pathways. These data indicate that vertical inhibition of proximal EGFR signaling should be pursued as a potential therapy to treat EGFR-mutated tumors. Lung cancer is the leading cause of cancer-related deaths worldwide. In non-smokers, this disease is usually caused by a mutation in a protein found on the surface of a cell, called EGFR. In healthy lung cells, these proteins trigger a chain of chemical signals that tell the cells to multiply. However, faulty forms of EFGR make the cells grow uncontrollably, leading to the formation of tumors. Current treatments use EGFR inhibitors that block the activity of these proteins. But cancer cells often become resistant to these treatments by activating other types of growth proteins. One way to overcome this resistance has been by targeting the signaling pathways within individual tumors. But since those pathways differ between tumors, it has been challenging to find a single therapy that can treat all drug-resistant cancer cells. Now, Theard et al. assessed the therapeutic effects of blocking a specific protein inside lung cells, called SOS1, which is involved in growth signaling in all tumor cells. Six different types of human lung cancer cells were used, all of which had faulty forms of EGFR, with three of the cell types showing drug resistance to current therapies. The cancer cells were either exposed to EGFR inhibitors only or to a combination of EGFR and SOS1 inhibitors. The most effective treatment was found to be through combinational therapy, with enhanced killing of drug-resistant cells. Theard et al. further assessed the effect of combinational therapy using cells kept in two different ways. Cancer cells were either grown in a two-dimensional format, with cells forming a single cell layer, or in a three-dimensional format, where cells were multi-layered and grew on top of each other as self-aggregating spheroids. Combinational therapy treatment was only successful when the cells where grown in a three-dimensional format. These findings highlight that future drug development studies should give consideration to the way cells are grown, as it can impact the results. They also provide a steppingstone towards tackling drug resistance in lung cancers that arise from EGFR mutations.
Collapse
Affiliation(s)
- Patricia L Theard
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - Erin Sheffels
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - Nancy E Sealover
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - Amanda J Linke
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - David J Pratico
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - Robert L Kortum
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, United States
| |
Collapse
|
19
|
Ong LJY, Zhu L, Tan GJS, Toh YC. Quantitative Image-Based Cell Viability (QuantICV) Assay for Microfluidic 3D Tissue Culture Applications. MICROMACHINES 2020; 11:mi11070669. [PMID: 32660019 PMCID: PMC7407956 DOI: 10.3390/mi11070669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 01/01/2023]
Abstract
Microfluidic 3D tissue culture systems are attractive for in vitro drug testing applications due to the ability of these platforms to generate 3D tissue models and perform drug testing at a very small scale. However, the minute cell number and liquid volume impose significant technical challenges to perform quantitative cell viability measurements using conventional colorimetric or fluorometric assays, such as MTS or Alamar Blue. Similarly, live-dead staining approaches often utilize metabolic dyes that typically label the cytoplasm of live cells, which makes it difficult to segment and count individual cells in compact 3D tissue cultures. In this paper, we present a quantitative image-based cell viability (QuantICV) assay technique that circumvents current challenges of performing the quantitative cell viability assay in microfluidic 3D tissue cultures. A pair of cell-impermeant nuclear dyes (EthD-1 and DAPI) were used to sequentially label the nuclei of necrotic and total cell populations, respectively. Confocal microscopy and image processing algorithms were employed to visualize and quantify the cell nuclei in the 3D tissue volume. The QuantICV assay was validated and showed good concordance with the conventional bulk MTS assay in static 2D and 3D tumor cell cultures. Finally, the QuantICV assay was employed as an on-chip readout to determine the differential dose responses of parental and metastatic 3D oral squamous cell carcinoma (OSCC) to Gefitinib in a microfluidic 3D culture device. This proposed technique can be useful in microfluidic cell cultures as well as in a situation where conventional cell viability assays are not available.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, #14-01, Singapore 117599, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Level 7, O Block, Gardens Point Campus, Brisbane City QLD 4000, Australia
| | - Liang Zhu
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Singapore Institute of Manufacturing Technology, 31 Biopolis Way, #04-10 Nanos, Singapore 138669, Singapore
- The N.1 Institute for Health, 28 Medical Drive, #05-corridor, Singapore 117456, Singapore
| | - Gabriel Jenn Sern Tan
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, #14-01, Singapore 117599, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Level 7, O Block, Gardens Point Campus, Brisbane City QLD 4000, Australia
- The N.1 Institute for Health, 28 Medical Drive, #05-corridor, Singapore 117456, Singapore
- NUS Tissue Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Q Block-IHBI, 60 Musk Avenue, Kelvin Grove QLD 4059, Australia
- Correspondence:
| |
Collapse
|
20
|
Sarne V, Huter S, Braunmueller S, Rakob L, Jacobi N, Kitzwögerer M, Wiesner C, Obrist P, Seeboeck R. Promoter Methylation of Selected Genes in Non-Small-Cell Lung Cancer Patients and Cell Lines. Int J Mol Sci 2020; 21:E4595. [PMID: 32605217 PMCID: PMC7369760 DOI: 10.3390/ijms21134595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 01/03/2023] Open
Abstract
Specific gene promoter DNA methylation is becoming a powerful epigenetic biomarker in cancer diagnostics. Five genes (CDH1, CDKN2Ap16, RASSF1A, TERT, and WT1) were selected based on their frequently published potential as epigenetic markers. Diagnostic promoter methylation assays were generated based on bisulfite-converted DNA pyrosequencing. The methylation patterns of 144 non-small-cell lung cancer (NSCLC) and 7 healthy control formalin-fixed paraffin-embedded (FFPE) samples were analyzed to evaluate the applicability of the putative diagnostic markers. Statistically significant changes in methylation levels are shown for TERT and WT1. Furthermore, 12 NSCLC and two benign lung cell lines were characterized for promoter methylation. The in vitro tests involved a comparison of promoter methylation in 2D and 3D cultures, as well as therapeutic tests investigating the impact of CDH1/CDKN2Ap16/RASSF1A/TERT/WT1 promoter methylation on sensitivity to tyrosine kinase inhibitor (TKI) and DNA methyl-transferase inhibitor (DNMTI) treatments. We conclude that the selected markers have potential and putative impacts as diagnostic or even predictive marker genes, although a closer examination of the resulting protein expression and pathway regulation is needed.
Collapse
MESH Headings
- Aged
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- DNA Methylation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Prognosis
- Promoter Regions, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Victoria Sarne
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Samuel Huter
- Pathologylab Dr. Obrist & Dr. Brunhuber OG, 6511 Zams, Austria; (S.H.); (P.O.)
| | - Sandrina Braunmueller
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Lisa Rakob
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Nico Jacobi
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Melitta Kitzwögerer
- Clinical Institute of Pathology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, 3100 St. Pölten, Austria;
| | - Christoph Wiesner
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
| | - Peter Obrist
- Pathologylab Dr. Obrist & Dr. Brunhuber OG, 6511 Zams, Austria; (S.H.); (P.O.)
| | - Rita Seeboeck
- Department Life Sciences, IMC University of Applied Sciences Krems, 3500 Krems, Austria; (V.S.); (S.B.); (L.R.); (N.J.); (C.W.)
- Clinical Institute of Pathology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, 3100 St. Pölten, Austria;
| |
Collapse
|
21
|
Brodwolf R, Volz-Rakebrand P, Stellmacher J, Wolff C, Unbehauen M, Haag R, Schäfer-Korting M, Zoschke C, Alexiev U. Faster, sharper, more precise: Automated Cluster-FLIM in preclinical testing directly identifies the intracellular fate of theranostics in live cells and tissue. Theranostics 2020; 10:6322-6336. [PMID: 32483455 PMCID: PMC7255044 DOI: 10.7150/thno.42581] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/01/2020] [Indexed: 12/25/2022] Open
Abstract
Fluorescence microscopy is widely used for high content screening in 2D cell cultures and 3D models. In particular, 3D tissue models are gaining major relevance in modern drug development. Enabling direct multiparametric evaluation of complex samples, fluorescence lifetime imaging (FLIM) adds a further level to intensity imaging by the sensitivity of the fluorescence lifetime to the microenvironment. However, the use of FLIM is limited amongst others by the acquisition of sufficient photon numbers without phototoxic effects in live cells. Herein, we developed a new cluster-based analysis method to enhance insight, and significantly speed up analysis and measurement time for the accurate translation of fluorescence lifetime information into pharmacological pathways. Methods: We applied a fluorescently-labeled dendritic core-multishell nanocarrier and its cargo Bodipy as molecules of interest (MOI) to human cells and reconstructed human tissue. Following the sensitivity and specificity assessment of the fitting-free Cluster-FLIM analysis of data in silico and in vitro, we evaluated the dynamics of cellular molecule uptake and intracellular interactions. For 3D live tissue investigations, we applied multiphoton (mp) FLIM. Owing to Cluster-FLIM's statistics-based fitting-free analysis, we utilized this approach for automatization. Results: To discriminate the fluorescence lifetime signatures of 5 different fluorescence species in a single color channel, the Cluster-FLIM method requires only 170, respectively, 90 counts per pixel to obtain 95% sensitivity (hit rate) and 95% specificity (correct rejection rate). Cluster-FLIM revealed cellular interactions of MOIs, representing their spatiotemporal intracellular fate. In a setting of an automated workflow, the assessment of lysosomal trapping of the MOI revealed relevant differences between normal and tumor cells, as well as between 2D and 3D models. Conclusion: The automated Cluster-FLIM tool is fitting-free, providing images with enhanced information, contrast, and spatial resolution at short exposure times and low fluorophore concentrations. Thereby, Cluster-FLIM increases the applicability of FLIM in high content analysis of target molecules in drug development and beyond.
Collapse
|
22
|
Popular three-dimensional models: Advantages for cancer, Alzheimer's and cardiovascular diseases. Tissue Cell 2020; 65:101367. [PMID: 32746982 DOI: 10.1016/j.tice.2020.101367] [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: 02/27/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 11/21/2022]
Abstract
The increasing prevalence of non-communicable diseases, namely cancer, Alzheimer's (AD) and cardiovascular diseases (CVDs), worldwide continues to be a major health burden. Research attempts have been made to understand the pathophysiology and develop effective therapeutic agents for these diseases using conventional in vitro and ex vivo models. Due to the complexity of human disease mechanisms, often these models fail to recapitulate clinically relevant pathologies. As such, interests are arising in the exploration of three-dimensional (3D) in-vitro models, which create an artificial environment to closely mimic in vivo human conditions. Several studies have developed 3D models for cancer, AD and CVD research which can greatly improve the understanding of biological mechanisms and mirror clinical drug activities. Thus, 3D cultures may provide new in-vitro models that recapitulate the architecture and biological mechanisms of human diseases prior to the need for the use of sentient animals.
Collapse
|
23
|
Modeling the Efficacy of Oncolytic Adenoviruses In Vitro and In Vivo: Current and Future Perspectives. Cancers (Basel) 2020; 12:cancers12030619. [PMID: 32155969 PMCID: PMC7139921 DOI: 10.3390/cancers12030619] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic adenoviruses (OAd) selectively target and lyse tumor cells and enhance anti- tumor immune responses. OAds have been used as promising cancer gene therapies for many years and there are a multitude of encouraging pre-clinical studies. However, translating OAd therapies to the clinic has had limited success, in part due to the lack of realistic pre-clinical models to rigorously test the efficacy of OAds. Solid tumors have a heterogenous and hostile microenvironment that provides many barriers to OAd treatment, including structural and immunosuppressive components that cannot be modeled in two-dimensional tissue culture. To replicate these characteristics and bridge the gap between pre-clinical and clinical success, studies must test OAd therapy in three-dimensional culture and animal models. This review focuses on current methods to test OAd efficacy in vitro and in vivo and the development of new model systems to test both oncolysis and immune stimulatory components of oncolytic adenovirotherapy.
Collapse
|
24
|
Santana-Codina N, Chandhoke AS, Yu Q, Małachowska B, Kuljanin M, Gikandi A, Stańczak M, Gableske S, Jedrychowski MP, Scott DA, Aguirre AJ, Fendler W, Gray NS, Mancias JD. Defining and Targeting Adaptations to Oncogenic KRASG12C Inhibition Using Quantitative Temporal Proteomics. Cell Rep 2020; 30:4584-4599.e4. [DOI: 10.1016/j.celrep.2020.03.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/04/2020] [Accepted: 03/07/2020] [Indexed: 02/07/2023] Open
|
25
|
Effects of microtubule-inhibiting small molecule and antibody-drug conjugate treatment on differentially-sized A431 squamous carcinoma spheroids. Sci Rep 2020; 10:907. [PMID: 31969631 PMCID: PMC6976639 DOI: 10.1038/s41598-020-57789-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Multicellular tumor spheroids have been increasingly used by researchers to produce more physiologically relevant experimental environments. However, tracking of spheroid growth and treatment-induced volume reduction has not been readily adopted. Here, squamous carcinoma cells were seeded at different starting cell numbers with growth and reduction kinetics monitored using live cell imaging. Following the initial growth phase, spheroids were treated with auristatin as small molecule (MMAE) or as antibody-drug conjugate containing non-cleavable auristatin drug payload (033-F). Compared to cells in monolayers, 033-F had notably weaker potency against spheroids despite potency levels of MMAE being similar against monolayers and spheroids. Accumulation of released payload from 033-F was reduced in higher volume spheroids, likely contributing to the potency differences. Despite lowered potency towards spheroids with 033-F, spheroid volume was still readily reduced by 033-F in a dose-dependent fashion, with >85% volume reductions at the highest concentrations for all spheroid sizes. Additionally, the core of the larger spheroids showed more resiliency towards microtubule inhibition. Overall, this work highlights how various in-vivo 'features' such as tumor penetration, cell interactions, and increased resistance to therapeutics can be integrated into a spheroid model and tracked over time by automated imaging technology.
Collapse
|
26
|
Baker AEG, Bahlmann LC, Tam RY, Liu JC, Ganesh AN, Mitrousis N, Marcellus R, Spears M, Bartlett JMS, Cescon DW, Bader GD, Shoichet MS. Benchmarking to the Gold Standard: Hyaluronan-Oxime Hydrogels Recapitulate Xenograft Models with In Vitro Breast Cancer Spheroid Culture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901166. [PMID: 31322299 DOI: 10.1002/adma.201901166] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/28/2019] [Indexed: 06/10/2023]
Abstract
Many 3D in vitro models induce breast cancer spheroid formation; however, this alone does not recapitulate the complex in vivo phenotype. To effectively screen therapeutics, it is urgently needed to validate in vitro cancer spheroid models against the gold standard of xenografts. A new oxime-crosslinked hyaluronan (HA) hydrogel is designed, manipulating gelation rate and mechanical properties to grow breast cancer spheroids in 3D. This HA-oxime breast cancer model maintains the gene expression profile most similar to that of tumor xenografts based on a pan-cancer gene expression profile (comprising 730 genes) of three different human breast cancer subtypes compared to Matrigel or conventional 2D culture. Differences in gene expression between breast cancer cultures in HA-oxime versus Matrigel or 2D are confirmed for 12 canonical pathways by gene set variation analysis. Importantly, drug response is dependent on the culture method. Breast cancer cells respond better to the Rac inhibitor (EHT-1864) and the PI3K inhibitor (AZD6482) when cultured in HA-oxime versus Matrigel. This study demonstrates the superiority of an HA-based hydrogel as a platform for in vitro breast cancer culture of both primary, patient-derived cells and cell lines, and provides a hydrogel culture model that closely matches that in vivo.
Collapse
Affiliation(s)
- Alexander E G Baker
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Laura C Bahlmann
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Roger Y Tam
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Jeffrey C Liu
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Ahil N Ganesh
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Nikolaos Mitrousis
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
| | - Richard Marcellus
- Ontario Institute for Cancer Research, MaRS Centre, 661 University Avenue, Toronto, Ontario, M5G 0A3, Canada
| | - Melanie Spears
- Ontario Institute for Cancer Research, MaRS Centre, 661 University Avenue, Toronto, Ontario, M5G 0A3, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - John M S Bartlett
- Ontario Institute for Cancer Research, MaRS Centre, 661 University Avenue, Toronto, Ontario, M5G 0A3, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, 610 University Ave., Toronto, Ontario, M5G 2C1, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Molly S Shoichet
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomaterials and Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
27
|
Black LE, Longo JF, Carroll SL. Mechanisms of Receptor Tyrosine-Protein Kinase ErbB-3 (ERBB3) Action in Human Neoplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1898-1912. [PMID: 31351986 DOI: 10.1016/j.ajpath.2019.06.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022]
Abstract
It is well established that the epidermal growth factor (EGF) receptor, receptor tyrosine-protein kinase erbB-2 (ERBB2)/human EGF receptor 2 (HER2), and, to a lesser extent, ERBB4/HER4, promote the pathogenesis of many types of human cancers. In contrast, the role that ERBB3/HER3, the fourth member of the ERBB family of receptor tyrosine kinases, plays in these diseases is poorly understood and, until recently, underappreciated. In large part, this was because early structural and functional studies suggested that ERBB3 had little, if any, intrinsic tyrosine kinase activity and, thus, was unlikely to be an important therapeutic target. Since then, however, numerous publications have demonstrated an important role for ERBB3 in carcinogenesis, metastasis, and acquired drug resistance. Furthermore, somatic ERBB3 mutations are frequently encountered in many types of human cancers. Dysregulation of ERBB3 trafficking as well as cooperation with other receptor tyrosine kinases further enhance ERBB3's role in tumorigenesis and drug resistance. As a result of these advances in our understanding of the structure and biochemistry of ERBB3, and a growing focus on the development of precision and combinatorial therapeutic regimens, ERBB3 is increasingly considered to be an important therapeutic target in human cancers. In this review, we discuss the unique structural and functional features of ERBB3 and how this information is being used to develop effective new therapeutic agents that target ERBB3 in human cancers.
Collapse
Affiliation(s)
- Laurel E Black
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Jody F Longo
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
28
|
Meier-Hubberten JC, Sanderson MP. Establishment and Analysis of a 3D Co-Culture Spheroid Model of Pancreatic Adenocarcinoma for Application in Drug Discovery. Methods Mol Biol 2019; 1953:163-179. [PMID: 30912022 DOI: 10.1007/978-1-4939-9145-7_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The high attrition rate of oncology drug candidates can be in part explained by the disconnect between the standard preclinical models (e.g., 2D culture, xenograft tumors) commonly employed for drug discovery and the complex multicellular microenvironment of human cancers. As such, significant focus has recently shifted to the establishment of preclinical models that more closely recapitulate human tumors, such as patient-derived xenografts, 3D spheroids, humanized mice, and mixed-culture models. For these models to be suited to drug discovery, they should optimally exhibit reproducibility, high-throughput, and robust and simple assay readouts. In this article, we describe a protocol for the generation of an in vitro 3D co-culture spheroid model that recapitulates the interaction of tumor cells with stromal fibroblasts in pancreatic adenocarcinoma. We additionally describe protocols relevant to the analysis of these spheroids in high-throughput drug discovery campaigns such as the assessment of spheroid proliferation, immunofluorescence and immunohistochemistry staining of spheroids, live-cell and confocal imaging and analysis of cell surface markers.
Collapse
|
29
|
Morales D, Lombart F, Truchot A, Maire P, Hussein M, Hamitou W, Vigneron P, Galmiche A, Lok C, Vayssade M. 3D Coculture Models Underline Metastatic Melanoma Cell Sensitivity to Vemurafenib. Tissue Eng Part A 2019; 25:1116-1126. [PMID: 30501565 DOI: 10.1089/ten.tea.2018.0210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
IMPACT STATEMENT Three dimensional in vitro cell culture systems better reflect the native structural architecture of tissues and are attractive to investigate cancer cell sensitivity to drugs. We have developed and compared several metastatic melanoma (MM) models cultured as a monolayer (2D) and cocultured on three dimensional (3D) dermal equivalents with fibroblasts to better unravel factors modulating cell sensitivity to vemurafenib, a BRAF inhibitor. The heterotypic 3D melanoma model we have established summarizes paracrine signalization by stromal cells and type I collagen matrix, mimicking the natural microenvironment of cutaneous MM, and allows for the identification of potent sensitive melanoma cells to the drug. This model could be a powerful tool for predicting drug efficiency.
Collapse
Affiliation(s)
- Delphine Morales
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Florian Lombart
- 2Department of Dermatology, CHU Amiens Picardie-Site Nord, Amiens, France
| | - Agathe Truchot
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Pauline Maire
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
- 3Department of Biochemistry, CHU Amiens Picardie, Amiens, France
| | - Marwa Hussein
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Warda Hamitou
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Pascale Vigneron
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| | - Antoine Galmiche
- 3Department of Biochemistry, CHU Amiens Picardie, Amiens, France
- 4Research Unit EA7516 CHIMERE, Université de Picardie Jules Verne, Amiens, France
| | - Catherine Lok
- 2Department of Dermatology, CHU Amiens Picardie-Site Nord, Amiens, France
| | - Muriel Vayssade
- 1Sorbonne University, Université de Technologie de Compiègne, CNRS, UMR 7338 Biomechanics and Bioengineering, Centre de Recherche Royallieu, Compiègne Cedex, France
| |
Collapse
|
30
|
Osswald A, Hedrich V, Sommergruber W. 3D-3 Tumor Models in Drug Discovery for Analysis of Immune Cell Infiltration. Methods Mol Biol 2019; 1953:151-162. [PMID: 30912021 DOI: 10.1007/978-1-4939-9145-7_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The cross talk between tumor cells and other cells present in the tumor microenvironment such as stromal and immune cells highly influences the behavior and progression of disease. Understanding the underlying mechanisms of interaction is a prerequisite to develop new treatment strategies and to prevent or at least reduce therapy failure in the future. Specific reactivation of the patient's immune system is one of the major goals today. However, standard two-dimensional (2D) cell culture techniques lack the necessary complexity to address related questions. Novel three-dimensional (3D) in vitro models-embedded in a matrix or encapsulated in alginate-recapitulate the in vivo situation much better. Cross talk between different cell types can be studied starting from co-cultures. As cancer immune modulation is becoming a major research topic, 3D in vitro models represent an important tool to address immune regulatory/modulatory questions for T, NK, and other cells of the immune system. The 3D systems consisting of tumor cells, fibroblasts, and immune cells (3D-3) already proved as a reliable tool for us. For instance, we made use of those models to study the molecular mechanisms of the cross talk of non-small cell lung cancer (NSCLC) and fibroblasts, to unveil macrophage plasticity in the tumor microenvironment and to mirror drug responses in vivo. Generation of those 3D models and how to use them to study immune cell infiltration and activation will be described in the present book chapter.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Bioreactors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cells, Immobilized/drug effects
- Cells, Immobilized/immunology
- Cells, Immobilized/pathology
- Coculture Techniques/methods
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/methods
- Fibroblasts/drug effects
- Fibroblasts/immunology
- Fibroblasts/pathology
- Humans
- Immunity, Cellular/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/immunology
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Viola Hedrich
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|