1
|
Guragain P, Singh S, Tavana H. Protocol to develop A 3D tumor model for drug testing applications. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 30:100206. [PMID: 39701269 DOI: 10.1016/j.slasd.2024.100206] [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: 08/31/2024] [Revised: 11/29/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Three-dimensional (3D) tumor models provide physiologically relevant tumor environments and have become a major tool in cancer research and drug discovery. This article presents a protocol for creating a 3D organotypic tumor model by embedding a cancer cell spheroid within a collagen matrix containing dispersed fibroblasts. This model offers significant advantages over the conventional monolayer cell culture, monoculture spheroids of cancer cells, and intermixed co-culture of cancer and stromal cells by mimicking the spatial organization and mechanical properties of a solid tumor. Compatible with robotic automation, our protocol significantly enhances reproducibility and scalability of creating a tumor model to study tumor-stromal interactions and test therapeutic compounds.
Collapse
Affiliation(s)
| | - Sunil Singh
- The University of Akron, Akron, OH, 44325, USA
| | | |
Collapse
|
2
|
Fries BD, Tobias F, Wang Y, Holbrook JH, Hummon AB. Lipidomics Profiling Reveals Differential Alterations after FAS Inhibition in 3D Colon Cancer Cell Culture Models. J Proteome Res 2024; 23:2919-2933. [PMID: 38063332 PMCID: PMC11161555 DOI: 10.1021/acs.jproteome.3c00593] [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] [Indexed: 03/21/2024]
Abstract
Cancerous cells synthesize most of their lipids de novo to keep up with their rapid growth and proliferation. Fatty acid synthase (FAS) is a key enzyme in the lipogenesis pathway that is upregulated in many cancers and has gained popularity as a druggable target of interest for cancer treatment. The first FAS inhibitor discovered, cerulenin, initially showed promise for chemotherapeutic purposes until it was observed that it had adverse side effects in mice. TVB-2640 (Denifanstat) is part of the newer generation of inhibitors. With multiple generations of FAS inhibitors being developed, it is vital to understand their distinct molecular downstream effects to elucidate potential interactions in the clinic. Here, we profile the lipidome of two different colorectal cancer (CRC) spheroids treated with a generation 1 inhibitor (cerulenin) or a generation 2 inhibitor (TVB-2640). We observe that the cerulenin causes drastic changes to the spheroid morphology as well as alterations to the lipid droplets found within CRC spheroids. TVB-2640 causes higher abundances of polyunsaturated fatty acids (PUFAs) whereas cerulenin causes a decreased abundance of PUFAs. The increase in PUFAs in TVB-2640 exposed spheroids indicates it is causing cells to die via a ferroptotic mechanism rather than a conventional apoptotic or necrotic mechanism.
Collapse
Affiliation(s)
- Brian D Fries
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Fernando Tobias
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Integrated Molecular Structure Education and Research Center (IMSERC), Northwestern University, Evanston, Illinois 60208, United States
| | - Yijia Wang
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Joseph H Holbrook
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio 43210, United States
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
3
|
Tovar-Parra D, Zammit-Mangion M. Comparative Analysis of the Effect of the BRAF Inhibitor Dabrafenib in 2D and 3D Cell Culture Models of Human Metastatic Melanoma Cells. In Vivo 2024; 38:1579-1593. [PMID: 38936891 PMCID: PMC11215570 DOI: 10.21873/invivo.13608] [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: 04/20/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM Melanoma, a variant of skin cancer, presents the highest mortality rates among all skin cancers. Despite advancements in targeted therapies, immunotherapies, and tissue culture techniques, the absence of an effective early treatment model remains a challenge. This study investigated the impact of dabrafenib on both 2D and 3D cell culture models with distinct molecular profiles. MATERIALS AND METHODS We developed a high-throughput workflow enabling drug screening on spheroids. Our approach involved cultivating 2D and 3D cultures derived from normal melanocytes and metastatic melanoma cells, treating them with dabrafenib and conducting viability, aggregation, migration, cell cycle, and apoptosis assays. RESULTS Dabrafenib exerted multifaceted influences, particularly on migration at concentrations of 10 and 25 μM. It induced a decrease in cell viability, impeded cellular adhesion to the matrix, inhibited cellular aggregation and spheroid formation, arrested the cell cycle in the G1 phase, and induced apoptosis. CONCLUSION These results confirm the therapeutic potential of dabrafenib in treating melanoma with the BRAF V600E mutation and that 3D models are validated models to study the potential of new molecules for therapeutic purposes. Furthermore, our study underscores the relevance of 3D models in simulating physiological in vivo microenvironments, providing insights into varied treatment responses between normal and tumor cells.
Collapse
Affiliation(s)
- David Tovar-Parra
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta;
| | - Marion Zammit-Mangion
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta;
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| |
Collapse
|
4
|
Coelho LL, Vianna MM, da Silva DM, Gonzaga BMDS, Ferreira RR, Monteiro AC, Bonomo AC, Manso PPDA, de Carvalho MA, Vargas FR, Garzoni LR. Spheroid Model of Mammary Tumor Cells: Epithelial-Mesenchymal Transition and Doxorubicin Response. BIOLOGY 2024; 13:463. [PMID: 39056658 PMCID: PMC11273983 DOI: 10.3390/biology13070463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 07/28/2024]
Abstract
Breast cancer is the most prevalent cancer among women worldwide. Therapeutic strategies to control tumors and metastasis are still challenging. Three-dimensional (3D) spheroid-type systems more accurately replicate the features of tumors in vivo, working as a better platform for performing therapeutic response analysis. This work aimed to characterize the epithelial-mesenchymal transition and doxorubicin (dox) response in a mammary tumor spheroid (MTS) model. We evaluated the doxorubicin treatment effect on MCF-7 spheroid diameter, cell viability, death, migration and proteins involved in the epithelial-mesenchymal transition (EMT) process. Spheroids were also produced from tumors formed from 4T1 and 67NR cell lines. MTSs mimicked avascular tumor characteristics, exhibited adherens junction proteins and independently produced their own extracellular matrix. Our spheroid model supports the 3D culturing of cells isolated from mice mammary tumors. Through the migration assay, we verified a reduction in E-cadherin expression and an increase in vimentin expression as the cells became more distant from spheroids. Dox promoted cytotoxicity in MTSs and inhibited cell migration and the EMT process. These results suggest, for the first time, that this model reproduces aspects of the EMT process and describes the potential of dox in inhibiting the metastatic process, which can be further explored.
Collapse
Affiliation(s)
- Laura Lacerda Coelho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Matheus Menezes Vianna
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Debora Moraes da Silva
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Beatriz Matheus de Souza Gonzaga
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Roberto Rodrigues Ferreira
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Ana Carolina Monteiro
- Laboratory of Osteo and Tumor Immunology, Department of Immunobiology, Fluminense Federal University (UFF), Rio de Janeiro 24020-150, Brazil;
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Adriana Cesar Bonomo
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Pedro Paulo de Abreu Manso
- Laboratory of Pathology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | | | - Fernando Regla Vargas
- Laboratory of Epidemiology of Congenital Malformations, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Luciana Ribeiro Garzoni
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| |
Collapse
|
5
|
Rahman MA, Apu EH, Rakib-Uz-Zaman SM, Chakraborti S, Bhajan SK, Taleb SA, Shaikh MH, Jalouli M, Harrath AH, Kim B. Exploring Importance and Regulation of Autophagy in Cancer Stem Cells and Stem Cell-Based Therapies. Cells 2024; 13:958. [PMID: 38891090 PMCID: PMC11171866 DOI: 10.3390/cells13110958] [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: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Autophagy is a globally conserved cellular activity that plays a critical role in maintaining cellular homeostasis through the breakdown and recycling of cellular constituents. In recent years, there has been much emphasis given to its complex role in cancer stem cells (CSCs) and stem cell treatment. This study examines the molecular processes that support autophagy and how it is regulated in the context of CSCs and stem cell treatment. Although autophagy plays a dual role in the management of CSCs, affecting their removal as well as their maintenance, the intricate interaction between the several signaling channels that control cellular survival and death as part of the molecular mechanism of autophagy has not been well elucidated. Given that CSCs have a role in the development, progression, and resistance to treatment of tumors, it is imperative to comprehend their biological activities. CSCs are important for cancer biology because they also show a tissue regeneration model that helps with organoid regeneration. In other words, the manipulation of autophagy is a viable therapeutic approach in the treatment of cancer and stem cell therapy. Both synthetic and natural substances that target autophagy pathways have demonstrated promise in improving stem cell-based therapies and eliminating CSCs. Nevertheless, there are difficulties associated with the limitations of autophagy in CSC regulation, including resistance mechanisms and off-target effects. Thus, the regulation of autophagy offers a versatile strategy for focusing on CSCs and enhancing the results of stem cell therapy. Therefore, understanding the complex interactions between autophagy and CSC biology would be essential for creating therapeutic treatments that work in both regenerative medicine and cancer treatment.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Global Biotechnology and Biomedical Research Network (GBBRN), Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA;
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - S. M Rakib-Uz-Zaman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Somdeepa Chakraborti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj 8100, Bangladesh;
| | - Shakila Afroz Taleb
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Mushfiq H. Shaikh
- Department of Otolaryngology—Head and Neck Surgery, Western University, London, ON N6A 4V2, Canada;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1-5 Hoegidong, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Lamichhane A, Tavana H. Three-Dimensional Tumor Models to Study Cancer Stemness-Mediated Drug Resistance. Cell Mol Bioeng 2024; 17:107-119. [PMID: 38737455 PMCID: PMC11082110 DOI: 10.1007/s12195-024-00798-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/01/2024] [Indexed: 05/14/2024] Open
Abstract
Solid tumors often contain genetically different populations of cancer cells, stromal cells, various structural and soluble proteins, and other soluble signaling molecules. The American Cancer society estimated 1,958,310 new cancer cases and 609,820 cancer deaths in the United States in 2023. A major barrier against successful treatment of cancer patients is drug resistance. Gain of stem cell-like states by cancer cells under drug pressure or due to interactions with the tumor microenvironment is a major mechanism that renders therapies ineffective. Identifying approaches to target cancer stem cells is expected to improve treatment outcomes for patients. Most of our understanding of drug resistance and the role of cancer stemness is from monolayer cell cultures. Recent advances in cell culture technologies have enabled developing sophisticated three-dimensional tumor models that facilitate mechanistic studies of cancer drug resistance. This review summarizes the role of cancer stemness in drug resistance and highlights the various tumor models that are used to discover the underlying mechanisms and test potentially novel therapeutics.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| |
Collapse
|
7
|
Gay MD, Drda JC, Chen W, Huang Y, Yassin AA, Duka T, Fang H, Shivapurkar N, Smith JP. Implicating the cholecystokinin B receptor in liver stem cell oncogenesis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G291-G309. [PMID: 38252699 PMCID: PMC11211039 DOI: 10.1152/ajpgi.00208.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related deaths worldwide. Chronic inflammation and fibrosis are the greatest risk factors for the development of HCC. Although the cell of origin for HCC is uncertain, many theories believe this cancer may arise from liver progenitor cells or stem cells. Here, we describe the activation of hepatic stem cells that overexpress the cholecystokinin-B receptor (CCK-BR) after liver injury with either a DDC diet (0.1% 3, 5-diethoxy-carbonyl 1,4-dihydrocollidine) or a NASH-inducing CDE diet (choline-deficient ethionine) in murine models. Pharmacologic blockade of the CCK-BR with a receptor antagonist proglumide or knockout of the CCK-BR in genetically engineered mice during the injury diet reduces the expression of hepatic stem cells and prevents the formation of three-dimensional tumorspheres in culture. RNA sequencing of livers from DDC-fed mice treated with proglumide or DDC-fed CCK-BR knockout mice showed downregulation of differentially expressed genes involved in cell proliferation and oncogenesis and upregulation of tumor suppressor genes compared with controls. Inhibition of the CCK-BR decreases hepatic transaminases, fibrosis, cytokine expression, and alters the hepatic immune cell signature rendering the liver microenvironment less oncogenic. Furthermore, proglumide hastened recovery after liver injury by reversing fibrosis and improving markers of synthetic function. Proglumide is an older drug that is orally bioavailable and being repurposed for liver conditions. These findings support a promising therapeutic intervention applicable to patients to prevent the development of HCC and decrease hepatic fibrosis.NEW & NOTEWORTHY This investigation identified a novel pathway involving the activation of hepatic stem cells and liver oncogenesis. Receptor blockade or genetic disruption of the cholecystokinin-B receptor (CCK-BR) signaling pathway decreased the activation and proliferation of hepatic stem cells after liver injury without eliminating the regenerative capacity of healthy hepatocytes.
Collapse
Affiliation(s)
- Martha D Gay
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Jack C Drda
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Wenqiang Chen
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Yimeng Huang
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| | - Amal A Yassin
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| | - Tetyana Duka
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Hongbin Fang
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, District of Columbia, United States
| | - Narayan Shivapurkar
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
| | - Jill P Smith
- Department of Medicine, Georgetown University, Washington, District of Columbia, United States
- Department of Oncology, Georgetown University, Washington, District of Columbia, United States
| |
Collapse
|
8
|
Xiong Q, Liu T, Ying Y, Yu X, Wang Z, Gao H, Lin T, Fan W, Zhang Z, Wei Q, Ge Y, Zeng S, Xu C. Establishment of bladder cancer spheroids and cultured in microfluidic platform for predicting drug response. Bioeng Transl Med 2024; 9:e10624. [PMID: 38435820 PMCID: PMC10905551 DOI: 10.1002/btm2.10624] [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: 03/23/2023] [Revised: 09/25/2023] [Accepted: 11/09/2023] [Indexed: 03/05/2024] Open
Abstract
Cisplatin-containing combination chemotherapy has been used as the standard treatment for bladder cancer patients at advanced stage. However, nearly 50% of patients are nonresponders. To guide the selection of more effective chemotherapeutic agents, a bladder cancer spheroids microfluidic drug sensitivity analysis system was established in this study. Bladder cancer spheroids were established and successfully cultured in a customized microfluidic device to assess their response to different chemotherapeutic agents. The in vitro drug sensitivity results were also compared to patient-derived xenograft (PDX) models and clinical responses of patients. As a result, bladder cancer spheroids faithfully recapitulate the histopathological and genetic features of their corresponding parental tumors. Furthermore, the in vitro drug sensitivity outcomes of spheroids (n = 8) demonstrated a high level of correlation with the PDX (n = 2) and clinical response in patients (n = 2). Our study highlights the potential of combining bladder cancer spheroids and microfluidic devices as an efficient and accurate platform for personalized selection of chemotherapeutic agents.
Collapse
Affiliation(s)
- Qiao Xiong
- Department of UrologyInstitute of Urology, West China Hospital, Sichuan UniversityChengduP. R. China
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Ting Liu
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information Technology, Chinese Academy of SciencesShanghaiP. R. China
| | - Yidie Ying
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Xiaowen Yu
- Department of GeriatricsChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Ziwei Wang
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Hongliang Gao
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Tianhai Lin
- Department of UrologyInstitute of Urology, West China Hospital, Sichuan UniversityChengduP. R. China
| | - Weihua Fan
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information Technology, Chinese Academy of SciencesShanghaiP. R. China
| | - Zhensheng Zhang
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Qiang Wei
- Department of UrologyInstitute of Urology, West China Hospital, Sichuan UniversityChengduP. R. China
| | - Yuqing Ge
- State Key Laboratory of Transducer TechnologyShanghai Institute of Microsystem and Information Technology, Chinese Academy of SciencesShanghaiP. R. China
| | - Shuxiong Zeng
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| | - Chuanliang Xu
- Department of UrologyChanghai Hospital, Naval Medical UniversityShanghaiP. R. China
| |
Collapse
|
9
|
Arutyunyan I, Jumaniyazova E, Makarov A, Fatkhudinov T. In Vitro Models of Head and Neck Cancer: From Primitive to Most Advanced. J Pers Med 2023; 13:1575. [PMID: 38003890 PMCID: PMC10672510 DOI: 10.3390/jpm13111575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
For several decades now, researchers have been trying to answer the demand of clinical oncologists to create an ideal preclinical model of head and neck squamous cell carcinoma (HNSCC) that is accessible, reproducible, and relevant. Over the past years, the development of cellular technologies has naturally allowed us to move from primitive short-lived primary 2D cell cultures to complex patient-derived 3D models that reproduce the cellular composition, architecture, mutational, or viral load of native tumor tissue. Depending on the tasks and capabilities, a scientific laboratory can choose from several types of models: primary cell cultures, immortalized cell lines, spheroids or heterospheroids, tissue engineering models, bioprinted models, organoids, tumor explants, and histocultures. HNSCC in vitro models make it possible to screen agents with potential antitumor activity, study the contribution of the tumor microenvironment to its progression and metastasis, determine the prognostic significance of individual biomarkers (including using genetic engineering methods), study the effect of viral infection on the pathogenesis of the disease, and adjust treatment tactics for a specific patient or groups of patients. Promising experimental results have created a scientific basis for the registration of several clinical studies using HNSCC in vitro models.
Collapse
Affiliation(s)
- Irina Arutyunyan
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov Ministry of Healthcare of the Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
| | - Andrey Makarov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Histology Department, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
10
|
Kulesza J, Paluszkiewicz E, Augustin E. Cellular Effects of Selected Unsymmetrical Bisacridines on the Multicellular Tumor Spheroids of HCT116 Colon and A549 Lung Cancer Cells in Comparison to Monolayer Cultures. Int J Mol Sci 2023; 24:15780. [PMID: 37958764 PMCID: PMC10649579 DOI: 10.3390/ijms242115780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Multicellular tumor spheroids are a good tool for testing new anticancer drugs, including those that may target cancer stem cells (CSCs), which are responsible for cancer progression, metastasis, and recurrence. Therefore, we applied this model in our studies of highly active antitumor unsymmetrical bisacridines (UAs). We investigated the cellular response induced by UAs in 2D and 3D cultures of HCT116 colon and A549 lung cancer cells, with an additional focus on their impact on the CSC-like population. We showed that UAs affected the viability of the studied cells, as well as their spherogenic potential in the 2D and 3D cultures. Furthermore, we proved that the most promising UAs (C-2045 and C-2053) induced apoptosis in the HCT116 and A549 spheres to a similar, or even higher, extent than what was found in monolayer conditions. Next, we identified the population of the CSC-like cells in the 2D and 3D cultures of the studied cell lines by determining the levels of CD166, CD133, CD44, and EpCAM markers. We showed that the selected UAs affected the CSC-like population in both of the cell lines, and that A549 was affected more profoundly in 3D than in 2D cultures. Thus, the UAs exhibited high antitumor properties in both the 2D and 3D conditions, which makes them promising candidates for future therapeutic applications.
Collapse
Affiliation(s)
| | | | - Ewa Augustin
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, 80-233 Gdańsk, Poland; (J.K.); (E.P.)
| |
Collapse
|
11
|
David KI, Ravikumar TS, Sethuraman S, Krishnan UM. Investigations of an organic-inorganic nanotheranostic hybrid for pancreatic cancer therapy using cancer-in-a-dish and in vivomodels. Biomed Mater 2022; 18. [PMID: 36270604 DOI: 10.1088/1748-605x/ac9cb2] [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: 11/19/2021] [Accepted: 10/21/2022] [Indexed: 12/14/2022]
Abstract
The incidence of highly aggressive pancreatic cancer is increasing across the globe and is projected to increase to 18.6% by 2050. The mortality rate for this form of cancer is very high and the 5 y relative survival rate is only about 9%-10%. The 3D pancreatic cancer microenvironment exerts a major influence on the poor survival rate. A key factor is the prevention of the penetration of the chemotherapeutic drugs in the three-dimensional (3D) microenvironment leading to the development of chemoresistance which is a major contributor to the survival rates. Hence,in vitrostudies using 3D cultures represent a better approach to understand the effect of therapeutic formulations on the cancer cells when compared to conventional 2D cultures. In the present study, we have explored three different conditions for the development of a 3D pancreatic tumour spheroid model from MiaPaCa-2 and PanC1 cells cultured for 10 days using Matrigel matrix. This optimized spheroid model was employed to evaluate a multi-functional nanotheranostic system fabricated using chitosan nanoparticles co-encapsulated with the chemotherapeutic agent gemcitabine and gold-capped iron oxide nanoparticles for multimodal imaging. The effect of the single and multiple-dose regimens of the theranostic system on the viability of 3D spheroids formed from the two pancreatic cancer cell lines was studied. It was observed that the 3D tumour spheroids cultured for 10 days exhibited resistance towards free gemcitabine drug, unlike the 2D culture. The administration of the multifunctional nanotheranostic system on alternate days effectively reduced the cancer cell viability after five doses to about 20% when compared with other groups. The repeated doses of the nanotheranostic system were found to be more effective than the single dose. Cell line-based differences in internalization of the carrier was also reflected in their response to the nanocarrier with PanC1 showing better sensitivity to the treatment.In vivostudies revealed that the combination of gemcitabine and magnetic field induced hypothermia produced superior regression in cancer when compared with the chemotherapeutic agent alone by a combination of activating the pro-apoptotic pathway and heat-induced necrosis. Our results reveal that this multi-functional system holds promise to overcome the current challenges to treat pancreatic cancers.
Collapse
Affiliation(s)
- Karolyn Infanta David
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| | - T S Ravikumar
- Formerly at Sri Venkateswara Institute of Medical Sciences (SVIMS) Tirupati 517507, India
| | - Swaminathan Sethuraman
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology and Advanced Biomaterials, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India.,School of Arts, Sciences, Humanities and Education, SASTRA Deemed University, Thanjavur, TamilNadu 613401, India
| |
Collapse
|
12
|
Ingavle G, Das M. Bench to Bedside: New Therapeutic Approaches with Extracellular Vesicles and Engineered Biomaterials for Targeting Therapeutic Resistance of Cancer Stem Cells. ACS Biomater Sci Eng 2022; 8:4673-4696. [PMID: 36194142 DOI: 10.1021/acsbiomaterials.2c00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer has recently been the second leading cause of death worldwide, trailing only cardiovascular disease. Cancer stem cells (CSCs), represented as tumor-initiating cells (TICs), are mainly liable for chemoresistance and disease relapse due to their self-renewal capability and differentiating capacity into different types of tumor cells. The intricate molecular mechanism is necessary to elucidate CSC's chemoresistance properties and cancer recurrence. Establishing efficient strategies for CSC maintenance and enrichment is essential to elucidate the mechanisms and properties of CSCs and CSC-related therapeutic measures. Current approaches are insufficient to mimic the in vivo chemical and physical conditions for the maintenance and growth of CSC and yield unreliable research results. Biomaterials are now widely used for simulating the bone marrow microenvironment. Biomaterial-based three-dimensional (3D) approaches for the enrichment of CSC provide an excellent promise for future drug discovery and elucidation of molecular mechanisms. In the future, the biomaterial-based model will contribute to a more operative and predictive CSC model for cancer therapy. Design strategies for materials, physicochemical cues, and morphology will offer a new direction for future modification and new methods for studying the CSC microenvironment and its chemoresistance property. This review highlights the critical roles of the microenvironmental cues that regulate CSC function and endow them with drug resistance properties. This review also explores the latest advancement and challenges in biomaterial-based scaffold structure for therapeutic approaches against CSC chemoresistance. Since the recent entry of extracellular vesicles (EVs), cell-derived nanostructures, have opened new avenues of investigation into this field, which, together with other more conventionally studied signaling pathways, play an important role in cell-to-cell communication. Thus, this review further explores the subject of EVs in-depth. This review also discusses possible future biomaterial and biomaterial-EV-based models that could be used to study the tumor microenvironment (TME) and will provide possible therapeutic approaches. Finally, this review concludes with potential perspectives and conclusions in this area.
Collapse
Affiliation(s)
- Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| | - Madhurima Das
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| |
Collapse
|
13
|
Sieber B, Lu F, Stribbling SM, Grieve AG, Ryan AJ, Freeman M. iRhom2 regulates ERBB signalling to promote KRAS-driven tumour growth of lung cancer cells. J Cell Sci 2022; 135:jcs259949. [PMID: 35971826 PMCID: PMC9482348 DOI: 10.1242/jcs.259949] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dysregulation of the ERBB/EGFR signalling pathway causes multiple types of cancer. Accordingly, ADAM17, the primary shedding enzyme that releases and activates ERBB ligands, is tightly regulated. It has recently become clear that iRhom proteins, inactive members of the rhomboid-like superfamily, are regulatory cofactors for ADAM17. Here, we show that oncogenic KRAS mutants target the cytoplasmic domain of iRhom2 (also known as RHBDF2) to induce ADAM17-dependent shedding and the release of ERBB ligands. Activation of ERK1/2 by oncogenic KRAS induces the phosphorylation of iRhom2, recruitment of the phospho-binding 14-3-3 proteins, and consequent ADAM17-dependent shedding of ERBB ligands. In addition, cancer-associated mutations in iRhom2 act as sensitisers in this pathway by further increasing KRAS-induced shedding of ERBB ligands. This mechanism is conserved in lung cancer cells, where iRhom activity is required for tumour xenograft growth. In this context, the activity of oncogenic KRAS is modulated by the iRhom2-dependent release of ERBB ligands, thus placing the cytoplasmic domain of iRhom2 as a central component of a positive feedback loop in lung cancer cells. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Boris Sieber
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Fangfang Lu
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - Adam G. Grieve
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Anderson J. Ryan
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Matthew Freeman
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
14
|
Xu Y, Pachnikova G, Wang H, Wu Y, Przybilla D, Schäfer R, Chen Z, Zhu S, Keilholz U. IC50: an unsuitable measure for large-sized prostate cancer spheroids in drug sensitivity evaluation. Bosn J Basic Med Sci 2022; 22:580-592. [PMID: 35694767 PMCID: PMC9392968 DOI: 10.17305/bjbms.2022.7279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/09/2022] [Indexed: 02/02/2023] Open
Abstract
Preclinical models of tumors have the potential to become valuable tools for commercial drug research and development, and 3D culture systems are gaining traction in this area, particularly in prostate cancer (PCa) research. However, nearly all 3D drug design and screening assessments are based on 2D experiments, suggesting limitations of 3D drug testing. To simulate the natural response of human cells to the drug, we detected the half-maximal inhibitory concentration (IC50) changes of 2D/3D LNCaP cells in the drug docetaxel, as well as the sensitivity of different morphologies of 2D/3D LNCaP to docetaxel treatment. In contrast to 2D LNCaP cells, the evaluation of LNCaP spheroids' susceptibility to treatment was more complicated; the fitness of IC50 curves of 2D and 3D tumor cell preclinical models differs significantly. IC50 curves were unsuitable for large-sized LNCaP spheroids. More evaluation indexes (such as max inhibition) and experiments (such as spheroids formation) should be explored and performed to evaluate the susceptibility systematically.
Collapse
Affiliation(s)
- Yipeng Xu
- Department of Urology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China,The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou, China,Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | - Gabriela Pachnikova
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - He Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Yaoyao Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Dorothea Przybilla
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Reinhold Schäfer
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Zihao Chen
- Department of Urology, Southern Medical University, Guangzhou, China
| | - Shaoxing Zhu
- Department of Urology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China,The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou, China,Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China,Corresponding authors: Shaoxing Zhu, Department of Urology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China; The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou, China; Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China. E-mail:
| | - Ulrich Keilholz
- Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany,German Cancer Consortium (DKTK), Heidelberg, Germany,
Ulrich Keilholz; Comprehensive Cancer Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Chariteplatz 1, 10117 Berlin, Germany; German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. E-mail:
| |
Collapse
|
15
|
Lipp C, Koebel L, Bertsch A, Gauthier M, Bolopion A, Renaud P. Dielectrophoretic Traps for Efficient Bead and Cell Trapping and Formation of Aggregates of Controlled Size and Composition. Front Bioeng Biotechnol 2022; 10:910578. [PMID: 35910025 PMCID: PMC9333130 DOI: 10.3389/fbioe.2022.910578] [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: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
We present a microfluidic dielectrophoretic-actuated system designed to trap chosen single-cell and form controlled cell aggregates. A novel method is proposed to characterize the efficiency of the dielectrophoretic trapping, considering the flow speed but also the heat generated by the traps as limiting criteria in cell-safe manipulation. Two original designs with different manufacturing processes are experimentally compared. The most efficient design is selected and the cell membrane integrity is monitored by fluorescence imaging to guarantee a safe-cell trapping. Design rules are suggested to adapt the traps to multiple-cells trapping and are experimentally validated as we formed aggregates of controlled size and composition with two different types of cells. We provide hereby a simple manufactured tool allowing the controlled manipulation of particles for the composition of multicellular assemblies.
Collapse
Affiliation(s)
- Clémentine Lipp
- Laboratory of Microsystems LMIS4, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Laure Koebel
- AS2M Department, CNRS, FEMTO-ST Institute, Université Bourgogne Franche-Comté, Besançon, France
| | - Arnaud Bertsch
- Laboratory of Microsystems LMIS4, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Michaël Gauthier
- AS2M Department, CNRS, FEMTO-ST Institute, Université Bourgogne Franche-Comté, Besançon, France
| | - Aude Bolopion
- AS2M Department, CNRS, FEMTO-ST Institute, Université Bourgogne Franche-Comté, Besançon, France
| | - Philippe Renaud
- Laboratory of Microsystems LMIS4, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- *Correspondence: Philippe Renaud,
| |
Collapse
|
16
|
Head T, Cady NC. Monitoring and modulation of the tumor microenvironment for enhanced cancer modeling. Exp Biol Med (Maywood) 2022; 247:598-613. [PMID: 35088603 PMCID: PMC9014523 DOI: 10.1177/15353702221074293] [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] [Indexed: 11/16/2022] Open
Abstract
Cancer treatments utilizing biologic or cytotoxic drugs compose the frontline of therapy, and though gains in treatment efficacy have been persistent in recent decades, much work remains in understanding cancer progression and treatment. Compounding this situation is the low rate of success when translating preclinical drug candidates to the clinic, which raises costs and development timelines. This underperformance is due in part to the poor recapitulation of the tumor microenvironment, a critical component of cancer biology, in cancer model systems. New technologies capable of both accurately observing and manipulating the tumor microenvironment are needed to effectively model cancer response to treatment. In this review, conventional cancer models are summarized, and a primer on emerging techniques for monitoring and modulating the tumor microenvironment is presented and discussed.
Collapse
Affiliation(s)
- Tristen Head
- College of Nanoscale Science & Engineering,
State University of New York Polytechnic Institute, Albany, NY 12203, USA
| | - Nathaniel C Cady
- College of Nanoscale Science & Engineering,
State University of New York Polytechnic Institute, Albany, NY 12203, USA
| |
Collapse
|
17
|
Mayer IM, Hoelbl-Kovacic A, Sexl V, Doma E. Isolation, Maintenance and Expansion of Adult Hematopoietic Stem/Progenitor Cells and Leukemic Stem Cells. Cancers (Basel) 2022; 14:1723. [PMID: 35406494 PMCID: PMC8996967 DOI: 10.3390/cancers14071723] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are rare, self-renewing cells that perch on top of the hematopoietic tree. The HSCs ensure the constant supply of mature blood cells in a tightly regulated process producing peripheral blood cells. Intense efforts are ongoing to optimize HSC engraftment as therapeutic strategy to treat patients suffering from hematopoietic diseases. Preclinical research paves the way by developing methods to maintain, manipulate and expand HSCs ex vivo to understand their regulation and molecular make-up. The generation of a sufficient number of transplantable HSCs is the Holy Grail for clinical therapy. Leukemia stem cells (LSCs) are characterized by their acquired stem cell characteristics and are responsible for disease initiation, progression, and relapse. We summarize efforts, that have been undertaken to increase the number of long-term (LT)-HSCs and to prevent differentiation towards committed progenitors in ex vivo culture. We provide an overview and compare methods currently available to isolate, maintain and enrich HSC subsets, progenitors and LSCs and discuss their individual advantages and drawbacks.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (I.M.M.); (A.H.-K.); (E.D.)
| | | |
Collapse
|
18
|
Langella A, Gadau SD, Serra E, Bebbere D, Ledda S. Microtubular Assessment of C6 Rat Glioma Cell Spheroids Developed in Transparent Liquid Marbles or Hanging Drops. BIOLOGY 2022; 11:biology11040492. [PMID: 35453692 PMCID: PMC9031767 DOI: 10.3390/biology11040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma is a brain tumour frequently used as an experimental model to exploit innovative therapeutic approaches due to its high lethality and refractoriness to therapies. Part of these innovative anticancer therapies address cytoskeletal microtubules (MTs) since specific tubulin post-translational modifications (PTMs) are considered markers of tumour plasticity. In vitro studies, which traditionally employ two-dimensional (2D) culture systems, are now being replaced by three-dimensional (3D) systems that more closely mimic in vivo physiological conditions and allow a better understanding of the signalling between cells. In this work, we compared 2 liquid base 3D methods for the generation of spheroids from C6 rat glioma cells (RGCs) using 30 µL of liquid marble (LM) or the hanging drops (HDs), which contained 2 different cell numbers (5000 or 15,000). After 24 or 48 h of in vitro culture (IVC), the morphology of the spheroids was observed and the behaviour of the two main tubulin PTMs, tyrosinated α-tubulin (Tyr-T) and acetylated α-tubulin (Ac-T), was evaluated by fluorescence and Western blot (WB). RGCs spontaneously formed spherical agglomerates more rapidly in the LM than in the HD system. Cell density influenced the size of the spheroids, which reached a larger size (> of 300 µm Ø), with 15,000 cells compared to 5000 cells (150 µm Ø). Moreover, an increase in Tyr-T and Ac-T was observed in both the HD and LM system from 24 to 48 h, with the highest values shown in the 48 h/LM spheroids of 5000 cells (p < 0.05). In conclusion, by comparing the morphology and microtubular architecture of spheroids from C6 rat glioma cells developed by LM or HD methodology, our findings demonstrate that the use of a fumed silica microbioreactor boosts the induction and maintenance of a high plasticity state in glioma cells. RGCs cultured in LM express levels of tubulin PTMs that can be used to evaluate the efficacy of new anticancer therapies.
Collapse
|
19
|
Chen Z, Han S, Sanny A, Chan DLK, van Noort D, Lim W, Tan AHM, Park S. 3D hanging spheroid plate for high-throughput CAR T cell cytotoxicity assay. J Nanobiotechnology 2022; 20:30. [PMID: 35012567 PMCID: PMC8744335 DOI: 10.1186/s12951-021-01213-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Background Most high-throughput screening (HTS) systems studying the cytotoxic effect of chimeric antigen receptor (CAR) T cells on tumor cells rely on two-dimensional cell culture that does not recapitulate the tumor microenvironment (TME). Tumor spheroids, however, can recapitulate the TME and have been used for cytotoxicity assays of CAR T cells. But a major obstacle to the use of tumor spheroids for cytotoxicity assays is the difficulty in separating unbound CAR T and dead tumor cells from spheroids. Here, we present a three-dimensional hanging spheroid plate (3DHSP), which facilitates the formation of spheroids and the separation of unbound and dead cells from spheroids during cytotoxicity assays. Results The 3DHSP is a 24-well plate, with each well composed of a hanging dripper, spheroid wells, and waste wells. In the dripper, a tumor spheroid was formed and mixed with CAR T cells. In the 3DHSP, droplets containing the spheroids were deposited into the spheroid separation well, where unbound and dead T and tumor cells were separated from the spheroid through a gap into the waste well by tilting the 3DHSP by more than 20°. Human epidermal growth factor receptor 2 (HER2)-positive tumor cells (BT474 and SKOV3) formed spheroids of approximately 300–350 μm in diameter after 2 days in the 3DHSP. The cytotoxic effects of T cells engineered to express CAR recognizing HER2 (HER2-CAR T cells) on these spheroids were directly measured by optical imaging, without the use of live/dead fluorescent staining of the cells. Our results suggest that the 3DHSP could be incorporated into a HTS system to screen for CARs that enable T cells to kill spheroids formed from a specific tumor type with high efficacy or for spheroids consisting of tumor types that can be killed efficiently by T cells bearing a specific CAR. Conclusions The results suggest that the 3DHSP could be incorporated into a HTS system for the cytotoxic effects of CAR T cells on tumor spheroids. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01213-8.
Collapse
Affiliation(s)
- Zhenzhong Chen
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
| | - Seokgyu Han
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
| | - Arleen Sanny
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Dorothy Leung-Kwan Chan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Danny van Noort
- Centro de Investigación en Bioingeniería, Universidad de Ingenieria y Tecnologia - UTEC, Lima 04, Peru.,Biotechnology, Linköping University, SE-581 83, Linköping, Sweden
| | - Wanyoung Lim
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
| | - Andy Hee-Meng Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore.
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea. .,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea. .,Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, South Korea.
| |
Collapse
|
20
|
OUP accepted manuscript. Glycobiology 2022; 32:588-599. [DOI: 10.1093/glycob/cwac016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/12/2022] Open
|
21
|
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
|
22
|
Aguilar Cosme JR, Gagui DC, Green NH, Bryant HE, Claeyssens F. In Vitro Low-Fluence Photodynamic Therapy Parameter Screening Using 3D Tumor Spheroids Shows that Fractionated Light Treatments Enhance Phototoxicity. ACS Biomater Sci Eng 2021; 7:5078-5089. [PMID: 34615346 DOI: 10.1021/acsbiomaterials.1c00690] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The evaluation of novel photosensitizers (PSs) for photodynamic therapy (PDT) is difficult due to the limitations of two-dimensional cell culture and multiple parameters (dose, light intensity, uptake time), which complicate progression to in vivo experiments and clinical translation. Three-dimensional (3D) cell culture models like multicellular cancer tumor spheroids (MCTS) show great similarities to in vivo avascular tumor conditions, improving the speed and accuracy of screening novel compounds with various treatment combinations. In this study, we utilize C8161 human melanoma spheroids to screen PDT treatment combinations using protoporphyrin IX (PpIX) and drug-loaded carbon dot (CD) conjugates PpIX-CD and PpIX@CD at ultralow fluence values (<10 J/cm2). Conjugates show equivalent light-induced damage to PpIX from 1 μg/mL with significantly less dark cytotoxicity up to 72 h after exposure, shown by LDH release and dsDNA content. Fractionated treatments, carried out by dividing light exposure with 24 h intervals, demonstrate an enhanced PDT effect compared to single exposure at equal concentrations. Light sheet fluorescence microscopy combined with live/dead staining demonstrates that spheroids sustain extensive damage after PDT, with PpIX and PpIX-CD showing improved uptake compared to PpIX@CD. We show that PDT parameter screening can be carried out using a low-cost and convenient combination of assays to improve the efficiency of evaluating novel compounds.
Collapse
Affiliation(s)
- Jose R Aguilar Cosme
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Dan C Gagui
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Nicola H Green
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| | - Helen E Bryant
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom
| | - Frederik Claeyssens
- University of Sheffield, Department of Materials Science and Engineering, Kroto Research Institute, Red Hill, Sheffield S3 7HQ, United Kingdom.,INSIGNEO Institute for In Silico Medicine, University of Sheffield, Pam Liversidge Building, Mappin Street, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
23
|
Porcheri C, Mitsiadis TA. New Scenarios in Pharmacological Treatments of Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2021; 13:cancers13215515. [PMID: 34771677 PMCID: PMC8583200 DOI: 10.3390/cancers13215515] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most frequent types of cancer with a lethal outcome in half of the diagnosed cases. Mostly, HNSCC develops in the oral cavity, and its development is associated with tobacco and areca nut/betel quid usage, alcohol consumption, and HPV infection. Oral squamous cell carcinoma, as other head and neck cancers, presents a high degree of intratumor heterogeneity, which makes their treatment difficult, and directly correlates with drug resistance. Since the classical treatments for HNSCC oftentimes do not resolve the clinical picture, there is great need for novel therapeutic approaches, models for drug testing, and new drug delivery systems.
Collapse
|
24
|
Dogan E, Kisim A, Bati-Ayaz G, Kubicek GJ, Pesen-Okvur D, Miri AK. Cancer Stem Cells in Tumor Modeling: Challenges and Future Directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2100017. [PMID: 34927168 PMCID: PMC8680587 DOI: 10.1002/anbr.202100017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microfluidic tumors-on-chips models have revolutionized anticancer therapeutic research by creating an ideal microenvironment for cancer cells. The tumor microenvironment (TME) includes various cell types and cancer stem cells (CSCs), which are postulated to regulate the growth, invasion, and migratory behavior of tumor cells. In this review, the biological niches of the TME and cancer cell behavior focusing on the behavior of CSCs are summarized. Conventional cancer models such as three-dimensional cultures and organoid models are reviewed. Opportunities for the incorporation of CSCs with tumors-on-chips are then discussed for creating tumor invasion models. Such models will represent a paradigm shift in the cancer community by allowing oncologists and clinicians to predict better which cancer patients will benefit from chemotherapy treatments.
Collapse
Affiliation(s)
- Elvan Dogan
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028
| | - Asli Kisim
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey
| | - Gizem Bati-Ayaz
- Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Gregory J. Kubicek
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, 2 Cooper Plaza, Camden, NJ 08103
| | - Devrim Pesen-Okvur
- Department of Molecular Biology & Genetics, Izmir Institute of Technology, Gulbahce Kampusu, Urla, Izmir, 35430, Turkey; Biotechnology and Bioengineering, Izmir Institute of Technology, Izmir, Turkey
| | - Amir K. Miri
- Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028; School of Medical Engineering, Science, and Health, Rowan University, Camden, NJ 08103
| |
Collapse
|
25
|
The Ultrastructural Analysis of Human Colorectal Cancer Stem Cell-Derived Spheroids and Their Mouse Xenograft Shows That the Same Cells Types Have Different Ratios. BIOLOGY 2021; 10:biology10090929. [PMID: 34571806 PMCID: PMC8465655 DOI: 10.3390/biology10090929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
Spheroids from primary colorectal cancer cells and their mice xenografts have emerged as useful preclinical models for cancer research as they replicate tumor features more faithfully as compared to cell lines. While 3D models provide a reliable system for drug discovery and testing, their structural complexity represents a challenge and their structure-function relationships are only partly understood. Here, we present a comparative ultrastructural and flow citometric analysis of patient colorectal cancer-derived spheroids and their mice xenografts. Ultrastructural observations highlighted that multicellular spheroids and their xenografts contain the same cancer cell types but with different ratios, specifically multicellular spheroids were enriched in cells with a stem-like phenotype, while xenografts had an increased amount of lipid droplets-containing cells. The flow cytometric analysis for stem cell marker and activity showed enrichment of stem-like cells presence and activity in spheroids while xenografts had the inverse response. Our results evidence the effects on cancer cells of different in vitro and in vivo microenvironments. Those differences have to be paid into account in designing innovative experimental models for personalized drug testing.
Collapse
|
26
|
She X, Gao Y, Zhao Y, Yin Y, Dong Z. A high-throughput screen identifies inhibitors of lung cancer stem cells. Biomed Pharmacother 2021; 140:111748. [PMID: 34044271 DOI: 10.1016/j.biopha.2021.111748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 01/02/2023] Open
Abstract
Metastasis is the main cause of cancer morbidity and mortality. Cancer stem cells (CSCs) are a rare subpopulation of cancer cells that can drive metastasis. The identification of CSC inhibitors and CSC-related genes is an alluring strategy for suppressing metastasis. Here, we established a simple and repeatable high-throughput CSC inhibitor screening platform that combined tumor sphere formation assays and cell viability assays. Human lung cancer cells were cocultured with 1280 pharmacologically active compounds (FDA-approved). Fifty-four candidate compounds obtained from our screening system completely or partially inhibited tumor sphere formation. A total of 5 of these 54 compounds (prochlorperazine dimaleate, thioridazine hydrochloride, ciproxifan hydrochloride, Ro 25-6981 hydrochloride, and AMN 082) completely inhibited the self-renewal of CSCs without cytotoxicity in vitro via their targets and suppressed lung cancer metastasis in vivo, suggesting that our screening platform is selective and reliable. DRD2, HRH3, and GRIN2B exhibited potent genes promoting CSCs in vitro experiments and clinical datasets. Further validation of the top hit (DRD2) and previously published studies demonstrate that our screening platform is a useful tool for CSC inhibitor and CSC-related gene screening.
Collapse
Affiliation(s)
- Xiaofei She
- School of Life Sciences and Technology, Cancer Center, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200092, China.
| | - Yaqun Gao
- School of Life Sciences and Technology, Cancer Center, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200092, China.
| | - Yan Zhao
- School of Life Sciences and Technology, Cancer Center, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200092, China
| | - Yue Yin
- School of Life Sciences and Technology, Cancer Center, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200092, China
| | - Zhewen Dong
- School of Life Sciences and Technology, Cancer Center, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200092, China
| |
Collapse
|
27
|
Tang LJW, Zaseela A, Toh CCM, Adine C, Aydar AO, Iyer NG, Fong ELS. Engineering stromal heterogeneity in cancer. Adv Drug Deliv Rev 2021; 175:113817. [PMID: 34087326 DOI: 10.1016/j.addr.2021.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 02/09/2023]
Abstract
Based on our exponentially increasing knowledge of stromal heterogeneity from advances in single-cell technologies, the notion that stromal cell types exist as a spectrum of unique subpopulations that have specific functions and spatial distributions in the tumor microenvironment has significant impact on tumor modeling for drug development and personalized drug testing. In this Review, we discuss the importance of incorporating stromal heterogeneity and tumor architecture, and propose an overall approach to guide the reconstruction of stromal heterogeneity in vitro for tumor modeling. These next-generation tumor models may support the development of more precise drugs targeting specific stromal cell subpopulations, as well as enable improved recapitulation of patient tumors in vitro for personalized drug testing.
Collapse
Affiliation(s)
- Leon Jia Wei Tang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Ayshath Zaseela
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | - Christabella Adine
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore
| | - Abdullah Omer Aydar
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - N Gopalakrishna Iyer
- National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, Singapore.
| | - Eliza Li Shan Fong
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore.
| |
Collapse
|
28
|
Koshkin V, De Oliveira MB, Peng C, Ailles LE, Liu G, Covens A, Krylov SN. Multi-drug-resistance efflux in cisplatin-naive and cisplatin-exposed A2780 ovarian cancer cells responds differently to cell culture dimensionality. Mol Clin Oncol 2021; 15:161. [PMID: 34295468 PMCID: PMC8273925 DOI: 10.3892/mco.2021.2323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/02/2021] [Indexed: 01/20/2023] Open
Abstract
A primary reason for chemotherapy failure is chemoresistance, which is driven by various mechanisms. Multi-drug resistance (MDR) is one such mechanism that is responsible for drug extrusion from the intracellular space. MDR can be intrinsic and thus, may pre-exist the first application of chemotherapy. However, MDR may also be acquired during tumor exposure to chemotherapeutic agents. To understand whether cell clustering can influence intrinsic and acquired MDR, the present study assessed cultured monolayers (representing individual cells) and spheroids (representing clusters) formed by cisplatin-naïve (intrinsic MDR) and cisplatin-exposed (acquired MDR) lines of ovarian cancer A2780 cells by determining the cytometry of reaction rate constant (CRRC). MDR efflux was characterized using accurate and robust cell number vs. MDR efflux rate constant (kMDR) histograms. Both cisplatin-naïve and cisplatin-exposed monolayer cells presented unimodal histograms; the histogram of cisplatin-exposed cells was shifted towards a higher kMDR value suggesting greater MDR activity. Spheroids of cisplatin-naïve cells presented a bimodal histogram indicating the presence of two subpopulations with different MDR activity. In contrast, spheroids of cisplatin-exposed cells presented a unimodal histogram qualitatively similar to that of the monolayers of cisplatin-exposed cells but with a moderate shift towards greater MDR activity. A flow-cytometry assessment of multidrug resistance-associated protein 1 transporter levels in monolayers and dissociated spheroids revealed distributions similar to those of kMDR, thus, suggesting a plausible molecular mechanism for the observed differences in MDR activity. The observed greater effect of cell clustering on intrinsic rather than in acquired MDR can help guide the development of new therapeutic strategies targeting clusters of circulating tumor cells.
Collapse
Affiliation(s)
- Vasilij Koshkin
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | | | - Chun Peng
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Laurie E Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Geoffrey Liu
- Department of Medicine, Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario M5G 2M9, Canada
| | - Allan Covens
- Sunnybrook Odette Cancer Centre, Toronto, Ontario M4N 3M5, Canada
| | - Sergey N Krylov
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
29
|
Lamichhane A, Thakuri PS, Rafsanjani Nejad P, Tavana H. Modeling adaptive drug resistance of colorectal cancer and therapeutic interventions with tumor spheroids. Exp Biol Med (Maywood) 2021; 246:2372-2380. [PMID: 34102903 DOI: 10.1177/15353702211014185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Drug resistance is a major barrier against successful treatments of cancer patients. Various intrinsic mechanisms and adaptive responses of tumor cells to cancer drugs often lead to failure of treatments and tumor relapse. Understanding mechanisms of cancer drug resistance is critical to develop effective treatments with sustained anti-tumor effects. Three-dimensional cultures of cancer cells known as spheroids present a biologically relevant model of avascular tumors and have been increasingly incorporated in tumor biology and cancer drug discovery studies. In this review, we discuss several recent studies from our group that utilized colorectal tumor spheroids to investigate responses of cancer cells to cytotoxic and molecularly targeted drugs and uncover mechanisms of drug resistance. We highlight our findings from both short-term, one-time treatments and long-term, cyclic treatments of tumor spheroids and discuss mechanisms of adaptation of cancer cells to the treatments. Guided by mechanisms of resistance, we demonstrate the feasibility of designing specific drug combinations to effectively block growth and resistance of cancer cells in spheroid cultures. Finally, we conclude with our perspectives on the utility of three-dimensional tumor models and their shortcomings and advantages for phenotypic and mechanistic studies of cancer drug resistance.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | | | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
30
|
Soeta K, Yamaguchi R, Iuchi K, Hisatomi H, Yokoyama C. Generation of Rat Monoclonal Antibody for Human IQGAP1 by Immunization of Three-Dimensional-Cultured Cancer Cells. Monoclon Antib Immunodiagn Immunother 2021; 40:118-123. [PMID: 34076498 DOI: 10.1089/mab.2020.0046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The scaffold protein IQ motif containing GTPase activating protein 1 (IQGAP1) is an adherens junction component in the epithelial tissue that binds many signaling and structural molecules to regulate biological processes. It is known that IQGAP1 is overexpressed in some tumors. In this study, we produced rat monoclonal antibodies (mAbs) through immunization of the lysate from three-dimensional (3D)-cultured DLD-1 cells to elucidate a characteristic feature of a tumor. In cancer research, 3D-cultured cancer cells are used as an intermediate model between in vitro cancer cell line cultures and in vivo tumors. Our results showed that mAb 7E11 recognized increasing antigen in the lysate of 3D-cultured cells comparing with two-dimensional-cultured cells, and its antigen is the human IQGAP1. Furthermore, we indicated that mAb 7E11 was used in immunoblotting, immunoprecipitation, and immunofluorescence staining. Therefore, it may be useful in the analysis of human cancer.
Collapse
Affiliation(s)
- Kenta Soeta
- Department of Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University, Yonezawa, Japan
| | - Rina Yamaguchi
- Department of Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University, Yonezawa, Japan
| | - Katsuya Iuchi
- Department of Materials and Life Science, Seikei University, Tokyo, Japan
| | - Hisashi Hisatomi
- Department of Materials and Life Science, Seikei University, Tokyo, Japan
| | - Chikako Yokoyama
- Department of Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University, Yonezawa, Japan
| |
Collapse
|
31
|
Abstract
Overcoming the challenges of understanding and treating cancer requires reliable patient-derived models of cancer (PDMCs). For decades, cancer research and therapeutic development relied primarily on cancer cell lines because of their prevalence, reproducibility, and simplicity to maintain. However, findings from research conducted in cell lines are rarely recapitulated in vivo and seldom directly translatable to patients. The tumor microenvironment (TME), tumor-stromal interactions, and associations with host immune cells produce profound changes in tumor phenotype and complexity not captured in traditional monolayer cell culture. In this chapter, we present various cancer explant models and discuss their applicability based on specific research aims. We discuss the appropriateness of these models for basic science questions, drug screening/development, and for personalized, precision medicine. We also consider logistical factors such as resource cost, technical difficulty, and accessibility. We finish this chapter with a practical guide intended to help the reader select the cancer explant model system(s) that best address their research aims.
Collapse
|
32
|
ADAM17 Inhibition Increases the Impact of Cisplatin Treatment in Ovarian Cancer Spheroids. Cancers (Basel) 2021; 13:cancers13092039. [PMID: 33922533 PMCID: PMC8122950 DOI: 10.3390/cancers13092039] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/15/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer (OvCa) treatment is still a challenge, mainly due to acquired resistance mechanisms during the course of chemotherapy. Here, we show the enhanced cytotoxicity of the combined treatment with the ADAM17 inhibitor GW280264X and cisplatin in comparison with cisplatin monotherapy. This effect was visible in five of five ovarian cancer cell lines grown as a monolayer and two of three tested cell lines in three-dimensional tumor spheroids. Tumor spheroids derived from primary tumor and ascites cells were sensitized to cisplatin treatment by GW280264X. In summary, the combination of ADAM17 inhibition with conventional chemotherapy seems to be a promising strategy to overcome chemotherapy resistance in OvCa. Abstract Chemotherapy resistance is a major challenge in ovarian cancer (OvCa). Thus, novel treatment combinations are highly warranted. However, many promising drug candidates tested in two-dimensional (2D) cell culture have not proved successful in the clinic. For this reason, we analyzed our drug combination not only in monolayers but also in three-dimensional (3D) tumor spheroids. One potential therapeutic target for OvCa is A disintegrin and metalloprotease 17 (ADAM17). ADAM17 can be activated by chemotherapeutics, which leads to enhanced tumor growth due to concomitant substrate cleavage. Therefore, blocking ADAM17 during chemotherapy may overcome resistance. Here, we tested the effect of the ADAM17 inhibitor GW280264X in combination with cisplatin on ovarian cancer cells in 2D and 3D. In 2D, the effect on five cell lines was analyzed with two readouts. Three of these cell lines formed dense aggregates or spheroids (HEY, SKOV-3, and OVCAR-8) in 3D and the treatment effect was analyzed with a multicontent readout (cytotoxicity, viability, and caspase3/7 activation). We tested the combined therapy on tumor spheroids derived from primary patient cells. In 2D, we found a significant reduction in the half minimal (50%) inhibitory concentration (IC50) value of the combined treatment (GW280264X plus cisplatin) in comparison with cisplatin monotherapy in all five cell lines with both 2D readout assays (viability and caspase activation). In contrast, the combined treatment only showed an IC50 reduction in HEY and OVCAR-8 3D tumor spheroid models using caspase3/7 activity or CelltoxTM Green as the readout. Finally, we found an improved effect of GW280264X with cisplatin in tumor spheroids derived from patient samples. In summary, we demonstrate that ADAM17 inhibition is a promising treatment strategy in ovarian cancer.
Collapse
|
33
|
Gel-Free 3D Tumoroids with Stem Cell Properties Modeling Drug Resistance to Cisplatin and Imatinib in Metastatic Colorectal Cancer. Cells 2021; 10:cells10020344. [PMID: 33562088 PMCID: PMC7914642 DOI: 10.3390/cells10020344] [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: 12/23/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 01/16/2023] Open
Abstract
Researchers have developed several three-dimensional (3D) culture systems, including spheroids, organoids, and tumoroids with increased properties of cancer stem cells (CSCs), also called cancer-initiating cells (CICs). Drug resistance is a crucial issue involving recurrence in cancer patients. Many studies on anti-cancer drugs have been reported using 2D culture systems, whereas 3D cultured tumoroids have many advantages for assessing drug sensitivity and resistance. Here, we aimed to investigate whether Cisplatin (a DNA crosslinker), Imatinib (a multiple tyrosine kinase inhibitor), and 5-Fluorouracil (5-FU: an antimetabolite) alter the tumoroid growth of metastatic colorectal cancer (mCRC). Gene expression signatures of highly metastatic aggregative CRC (LuM1 cells) vs. low-metastatic, non-aggregative CRC (Colon26 and NM11 cells) were analyzed using microarray. To establish a 3D culture-based multiplexing reporter assay system, LuM1 was stably transfected with the Mmp9 promoter-driven ZsGreen fluorescence reporter gene, which was designated as LuM1/m9 cells and cultured in NanoCulture Plate®, a gel-free 3D culture device. LuM1 cells highly expressed mRNA encoding ABCG2 (a drug resistance pump, i.e., CSC/CIC marker), other CSC/CIC markers (DLL1, EpCAM, podoplanin, STAT3/5), pluripotent stem cell markers (Sox4/7, N-myc, GATA3, Nanog), and metastatic markers (MMPs, Integrins, EGFR), compared to the other two cell types. Hoechst efflux stem cell-like side population was increased in LuM1 (7.8%) compared with Colon26 (2.9%), both of which were markedly reduced by verapamil treatment, an ABCG2 inhibitor. Smaller cell aggregates of LuM1 were more sensitive to Cisplatin (at 10 μM), whereas larger tumoroids with increased ABCG2 expression were insensitive. Notably, Cisplatin (2 μM) and Imatinib (10 μM) at low concentrations significantly promoted tumoroid formation (cell aggregation) and increased Mmp9 promoter activity in mCRC LuM1/m9, while not cytotoxic to them. On the other hand, 5-FU significantly inhibited tumoroid growth, although not completely. Thus, drug resistance in cancer with increased stem cell properties was modeled using the gel-free 3D cultured tumoroid system. The tumoroid culture is useful and easily accessible for the assessment of drug sensitivity and resistance.
Collapse
|
34
|
Abstract
Sphere assays are widely used in vitro techniques to enrich and evaluate the stem-like cell behavior of both normal and cancer cells. Utilizing three-dimensional in vitro sphere culture conditions provide a better representation of tumor growth in vivo than the more common monolayer cultures. We describe how to perform primary and secondary sphere assays, used for the enrichment and self-renewability studies of melanoma/melanocyte stem-like cells. Spheres are generated by growing melanoma cells at low density in nonadherent conditions with stem cell media. We provide protocols for preparing inexpensive and versatile polyHEMA-coated plates, setting up primary and secondary sphere assays in almost any tissue culture format and quantification methods using standard inverted microscopy. Our protocol is easily adaptable to laboratories with basic cell culture capabilities, without the need for expensive fluidic instruments.
Collapse
|
35
|
Chen Z, Wang F, Zhang J, Sun X, Yan Y, Wang Y, Ouyang J, Zhang J, Honore T, Ge J, Gu Z. Study on Development of Composite Hydrogels With Tunable Structures and Properties for Tumor-on-a-Chip Research. Front Bioeng Biotechnol 2020; 8:611796. [PMID: 33425875 PMCID: PMC7786431 DOI: 10.3389/fbioe.2020.611796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
A major factor for developing new tumor models is to recreate a proper three-dimensional environment for 3D tumors culture. In this 3D microenvironment, extracellular matrices play important roles in regulation of hallmark features of cancer through biochemical and mechanical signals. The fabrication of a mechanical and biophysical controllable hydrogel, while sharing similarities with Matrigel in cancer invasiveness evaluation, is an urgent but unmet need. In this study, we developed a hybrid hydrogel system composed of GelMA and hydrolyzed collagen to model tumor micro-environment and tested with several cancer cells with different origin and characteristics. This hydrogel possesses a well-ordered homogenous microstructure, excellent permeability and an adjustable mechanical stiffness. This hydrogel demonstrated similar properties as Matrigel in tumor spheroids culture and 3D tumor invasiveness studies. It was further applied in a Tumor-on-a-Chip system with 3D-bioprinting. Our research demonstrated this hydrogel's effectiveness in tumor 3D culture, and its potential to replace Matrigel in cancer invasiveness evaluation.
Collapse
Affiliation(s)
- Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Fei Wang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Jie Zhang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Xiaowei Sun
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Yuchuan Yan
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Yan Wang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Jun Ouyang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Jing Zhang
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Tess Honore
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Jianjun Ge
- Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
36
|
LaValley DJ, Miller PG, Shuler ML. Pumpless, unidirectional microphysiological system for testing metabolism-dependent chemotherapeutic toxicity. Biotechnol Prog 2020; 37:e3105. [PMID: 33274840 DOI: 10.1002/btpr.3105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022]
Abstract
Drug development is often hindered by the failure of preclinical models to accurately assess and predict the efficacy and safety of drug candidates. Body-on-a-chip (BOC) microfluidic devices, a subset of microphysiological systems (MPS), are being created to better predict human responses to drugs. Each BOC is designed with separate organ chambers interconnected with microfluidic channels mimicking blood recirculation. Here, we describe the design of the first pumpless, unidirectional, multiorgan system and apply this design concept for testing anticancer drug treatments. HCT-116 colon cancer spheroids, HepG2/C3A hepatocytes, and HL-60 promyeloblasts were embedded in collagen hydrogels and cultured within compartments representing "colon tumor", "liver," and "bone marrow" tissue, respectively. Operating on a pumpless platform, the microfluidic channel design provides unidirectional perfusion at physiologically realistic ratios to multiple channels simultaneously. The metabolism-dependent toxic effect of Tegafur, an oral prodrug of 5-fluorouracil, combined with uracil was examined in each cell type. Tegafur-uracil treatment induced substantial cell death in HCT-116 cells and this cytotoxic response was reduced for multicellular spheroids compared to single cells, likely due to diffusion-limited drug penetration. Additionally, off-target toxicity was detected by HL-60 cells, which demonstrate that such systems can provide useful information on dose-limiting side effects. Collectively, this microscale cell culture analog is a valuable physiologically-based pharmacokinetic drug screening platform that may be used to support cancer drug development.
Collapse
Affiliation(s)
- Danielle J LaValley
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Paula G Miller
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA.,Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
37
|
Mao Y, Zhao Y, Zhang Y, Yang H. In-depth characterization and comparison of the N-glycosylated proteome of two-dimensional- and three-dimensional-cultured breast cancer cells and xenografted tumors. PLoS One 2020; 15:e0243789. [PMID: 33301522 PMCID: PMC7728280 DOI: 10.1371/journal.pone.0243789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/25/2020] [Indexed: 02/05/2023] Open
Abstract
Native intact N-glycopeptide analysis can provide access to the comprehensive characteristics of N-glycan occupancy, including N-glycosites, N-glycan compositions, and N-glycoproteins for complex samples. The sample pre-processing method used for the analysis of intact N-glycopeptides usually depends on the enrichment of low abundance N-glycopeptides from a tryptic peptide mixture using hydrophilic substances before LC-MS/MS detection. However, the number of identified intact N-glycopeptides remains inadequate to achieve an in-depth profile of the N-glycosylation landscape. Here, we optimized the sample preparation workflow prior to LC-MS/MS analysis by systematically comparing different analytical methods, including the use of different sources of trypsin, combinations of different proteases, and different enrichment materials. Finally, we found that the combination of Trypsin (B)/Lys-C digestion and zwitterionic HILIC (Zic-HILIC) enrichment significantly improved the mass spectrometric characterization of intact N-glycopeptides, increasing the number of identified intact N-glycopeptides and offering better analytical reproducibility. Furthermore, the optimized workflow was applied to the analysis of intact N-glycopeptides in two-dimensional (2D) and three-dimensional (3D)-cultured breast cancer cells in vitro and xenografted tumors in mice. These results indicated that the same breast cancer cells, when cultured in different microenvironments, can show different N-glycosylation patterns. This study also provides an interesting comparison of the N-glycoproteome of breast cancer cells cultured in different growth conditions, indicating the important role of N-glycosylated proteins in cancer cell growth and the choice of the cell culture model for studies in tumor biology and drug evaluation.
Collapse
Affiliation(s)
- Yonghong Mao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Zhao
- Mass Spectrometry Engineering Technology Research Center, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Yong Zhang
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University, Chengdu, China
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- * E-mail: (YZ); (HY)
| | - Hao Yang
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University, Chengdu, China
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- * E-mail: (YZ); (HY)
| |
Collapse
|
38
|
Cucchi DGJ, Groen RWJ, Janssen JJWM, Cloos J. Ex vivo cultures and drug testing of primary acute myeloid leukemia samples: Current techniques and implications for experimental design and outcome. Drug Resist Updat 2020; 53:100730. [PMID: 33096284 DOI: 10.1016/j.drup.2020.100730] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
New treatment options of acute myeloid leukemia (AML) are rapidly emerging. Pre-clinical models such as ex vivo cultures are extensively used towards the development of novel drugs and to study synergistic drug combinations, as well as to discover biomarkers for both drug response and anti-cancer drug resistance. Although these approaches empower efficient investigation of multiple drugs in a multitude of primary AML samples, their translational value and reproducibility are hampered by the lack of standardized methodologies and by culture system-specific behavior of AML cells and chemotherapeutic drugs. Moreover, distinct research questions require specific methods which rely on specific technical knowledge and skills. To address these aspects, we herein review commonly used culture techniques in light of diverse research questions. In addition, culture-dependent effects on drug resistance towards commonly used drugs in the treatment of AML are summarized including several pitfalls that may arise because of culture technique artifacts. The primary aim of the current review is to provide practical guidelines for ex vivo primary AML culture experimental design.
Collapse
Affiliation(s)
- D G J Cucchi
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - R W J Groen
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - J J W M Janssen
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - J Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands.
| |
Collapse
|
39
|
Shahi Thakuri P, Lamichhane A, Singh S, Gupta M, Luker GD, Tavana H. Modeling Adaptive Resistance of KRAS Mutant Colorectal Cancer to MAPK Pathway Inhibitors with a Three-Dimensional Tumor Model. ACS Pharmacol Transl Sci 2020; 3:1176-1187. [PMID: 33344895 DOI: 10.1021/acsptsci.0c00115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Indexed: 02/07/2023]
Abstract
Single-agent drug treatment of KRASmut colorectal cancers is often ineffective because the activation of compensatory signaling pathways leads to drug resistance. To mimic cyclic chemotherapy treatments of patients, we showed that intermittent treatments of 3D tumor spheroids of KRASmut colorectal cancer cells with inhibitors of mitogen-activated protein kinase (MAPK) signaling pathway temporarily suppressed growth of spheroids. However, the efficacy of successive single-agent treatments was significantly reduced. Molecular analysis showed compensatory activation of PI3K/AKT and STAT kinases and EGFR family proteins. To overcome the adaptation of cancer cells to MAPK pathway inhibitors, we treated tumor spheroids with a combination of MEK and EGFR inhibitors. This approach significantly blocked signaling of MAPK and PI3K/AKT pathways and prevented the growth of spheroids, but it was not effective against STAT signaling. Although the combination treatment blocked the matrix invasion of DLD1 cells, additional treatments with STAT inhibitors were necessary to prevent invasiveness of HCT116 cells. Overall, our drug resistance model elucidated the mechanisms of treatment-induced growth and invasiveness of cancer cells and allowed design-driven testing and identifying of effective treatments to suppress these phenotypes.
Collapse
Affiliation(s)
- Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Sunil Singh
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Megha Gupta
- Department of Arts and Sciences, The University of Akron, Akron, Ohio 44325, United States
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States.,Department of Radiology, Microbiology and Immunology, and Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States.,Department of Radiology, Microbiology and Immunology, and Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
40
|
Gilazieva Z, Ponomarev A, Rutland C, Rizvanov A, Solovyeva V. Promising Applications of Tumor Spheroids and Organoids for Personalized Medicine. Cancers (Basel) 2020; 12:E2727. [PMID: 32977530 PMCID: PMC7598156 DOI: 10.3390/cancers12102727] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
One of the promising directions in personalized medicine is the use of three-dimensional (3D) tumor models such as spheroids and organoids. Spheroids and organoids are three-dimensional cultures of tumor cells that can be obtained from patient tissue and, using high-throughput personalized medicine methods, provide a suitable therapy for that patient. These 3D models can be obtained from most types of tumors, which provides opportunities for the creation of biobanks with appropriate patient materials that can be used to screen drugs and facilitate the development of therapeutic agents. It should be noted that the use of spheroids and organoids would expand the understanding of tumor biology and its microenvironment, help develop new in vitro platforms for drug testing and create new therapeutic strategies. In this review, we discuss 3D tumor spheroid and organoid models, their advantages and disadvantages, and evaluate their promising use in personalized medicine.
Collapse
Affiliation(s)
- Zarema Gilazieva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Z.G.); (A.P.); (A.R.)
| | - Aleksei Ponomarev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Z.G.); (A.P.); (A.R.)
| | - Catrin Rutland
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Z.G.); (A.P.); (A.R.)
| | - Valeriya Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Z.G.); (A.P.); (A.R.)
| |
Collapse
|
41
|
Head and Neck Cancer Stem Cell-Enriched Spheroid Model for Anticancer Compound Screening. Cells 2020; 9:cells9071707. [PMID: 32708734 PMCID: PMC7408407 DOI: 10.3390/cells9071707] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs), a rare cell population in tumors, are resistant to conventional chemotherapy and thus responsible for tumor recurrence. To screen for active compounds targeting CSCs, a good CSC-enriched model compatible with high-throughput screening (HTS) is needed. Here, we describe a new head and neck cancer stem cell-enriched spheroid model (SCESM) suitable for HTS analyses of anti-CSC compounds. We used FaDu cells, round-bottom ultra-low adherent (ULA) microplates, and stem medium. The formed spheroids displayed increased expression of all stem markers tested (qRT-PCR and protein analysis) in comparison to the FaDu cells grown in a standard adherent culture or in a well-known HTS-compatible multi-cellular tumor spheroid model (MCTS). Consistent with increased stemness of the cells in the spheroid, confocal microscopy detected fast proliferating cells only at the outer rim of the SCESM spheroids, with poorly/non-proliferating cells deeper in. To confirm the sensitivity of our model, we used ATRA treatment, which strongly reduced the expression of selected stem markers. Altogether, we developed a CSC-enriched spheroid model with a simple protocol, a microplate format compatible with multimodal detection systems, and a high detection signal, making it suitable for anti-CSC compounds' HTS.
Collapse
|
42
|
Zhang C, Yang Z, Dong DL, Jang TS, Knowles JC, Kim HW, Jin GZ, Xuan Y. 3D culture technologies of cancer stem cells: promising ex vivo tumor models. J Tissue Eng 2020; 11:2041731420933407. [PMID: 32637062 PMCID: PMC7318804 DOI: 10.1177/2041731420933407] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer stem cells have been shown to be important in tumorigenesis processes, such as tumor growth, metastasis, and recurrence. As such, many three-dimensional models have been developed to establish an ex vivo microenvironment that cancer stem cells experience under in vivo conditions. Cancer stem cells propagating in three-dimensional culture systems show physiologically related signaling pathway profiles, gene expression, cell-matrix and cell-cell interactions, and drug resistance that reflect at least some of the tumor properties seen in vivo. Herein, we discussed the presently available Cancer stem cell three-dimensional culture models that use biomaterials and engineering tools and the biological implications of these models compared to the conventional ones.
Collapse
Affiliation(s)
- Chengye Zhang
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji, China.,Air Force Medical Center of the Chinese PLA, Beijing, China
| | - Zhaoting Yang
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji, China
| | - Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Tae-Su Jang
- Department of Pre-Medical Course, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Yanhua Xuan
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji, China.,Department of Pathology, Yanbian University College of Medicine, Yanji, China
| |
Collapse
|
43
|
Fiorentzis M, Katopodis P, Kalirai H, Seitz B, Viestenz A, Coupland SE. Image Analysis of 3D Conjunctival Melanoma Cell Cultures Following Electrochemotherapy. Biomedicines 2020; 8:biomedicines8060158. [PMID: 32545782 PMCID: PMC7344416 DOI: 10.3390/biomedicines8060158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) cell cultures represent small avascular tumors in vitro and simulate some of the biological characteristics of solid tumors, enhancing the evaluation of anticancer drug efficacy. Automated image analysis can be used for the assessment of tumor growth and documentation of changes in the size parameters of 3D tumor spheroids following anticancer treatments such as electrochemotherapy. The objective of this article is to assess the effect of various electroporation (EP) conditions (500-750 Volts/cm, 8-20 pulses, 100 µs pulse duration, 5 Hz repetition rate) combined with different bleomycin concentrations (1-2.5 ug/mL) on normal epithelial (HCjE-Gi) and conjunctival melanoma (CRMM1, CRMM2) 3D-cell cultures, through an automated image analysis and a comparison with standard histological assays. A reduction in tumor mass with loss of cell definition was observed after ECT (750 Volts/cm with eight pulses and 500 Volts/cm with 20 pulses) with bleomycin (1 μg/mL and 2.5 μg/mL) in the histological and immunohistochemical analyses of 3D CRMM1 and CRMM2 spheroids, whereas an increase in volume and a decrease in sphericity was documented in the automated image analysis and 3D visualization of both melanoma cell lines. For all other treatment conditions and for the HCjE-Gi cell line, no significant changes to their morphological features were observed. Image analysis with integrated software tools provides an accessible and comprehensive platform for the preliminary selection of homogenous spheroids and for the monitoring of drug efficacy, implementing the traditional screening methods.
Collapse
Affiliation(s)
- Miltiadis Fiorentzis
- Department of Ophthalmology, University Hospital Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-2900
| | - Periklis Katopodis
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University, London UB8 3PH, UK;
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK; (H.K.); (S.E.C.)
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool L69 3GA, UK
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, 66424 Homburg, Germany;
| | - Arne Viestenz
- Department of Ophthalmology, University Hospital Halle, 06112 Halle, Germany;
| | - Sarah E. Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK; (H.K.); (S.E.C.)
- Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool L69 3GA, UK
| |
Collapse
|
44
|
Wodzanowski KA, Cassel SE, Grimes CL, Kloxin AM. Tools for probing host-bacteria interactions in the gut microenvironment: From molecular to cellular levels. Bioorg Med Chem Lett 2020; 30:127116. [PMID: 32223923 PMCID: PMC7476074 DOI: 10.1016/j.bmcl.2020.127116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/28/2020] [Accepted: 03/15/2020] [Indexed: 12/31/2022]
Abstract
Healthy function of the gut microenvironment is dependent on complex interactions between the bacteria of the microbiome, epithelial and immune (host) cells, and the surrounding tissue. Misregulation of these interactions is implicated in disease. A range of tools have been developed to study these interactions, from mechanistic studies to therapeutic evaluation. In this Digest, we highlight select tools at the cellular and molecular level for probing specific cell-microenvironment interactions. Approaches are overviewed for controlling and probing cell-cell interactions, from transwell and microfluidic devices to engineered bacterial peptidoglycan fragments, and cell-matrix interactions, from three-dimensional scaffolds to chemical handles for in situ modifications.
Collapse
Affiliation(s)
| | - Samantha E Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Catherine L Grimes
- Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, United States; Biological Sciences, University of Delaware, Newark, DE 19716, United States.
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States; Materials Science and Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
45
|
Thakuri PS, Gupta M, Plaster M, Tavana H. Quantitative Size-Based Analysis of Tumor Spheroids and Responses to Therapeutics. Assay Drug Dev Technol 2020; 17:140-149. [PMID: 30958703 DOI: 10.1089/adt.2018.895] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Drug resistance remains a major clinical problem despite advances in targeted therapies. In recent years, methods to culture cancer cells in three-dimensional (3D) environments to better mimic native tumors have gained increasing popularity. Nevertheless, unlike traditional two-dimensional (2D) cell cultures, analysis of 3D cultures is not straightforward. Most biochemical assays developed for 2D cultures have to be optimized for use with 3D cultures. We addressed this important problem by presenting a simple method of quantitative size-based analysis of growth and drug responses of 3D cultures of cancer cells as tumor spheroids. We used an aqueous two-phase system to form consistently sized tumor spheroids of colorectal cancer cells. Using spheroid images, we computed the size of spheroids over time and demonstrated that growth of spheroids from this analysis strongly correlates with that using a PrestoBlue biochemical assay optimized for 3D cultures. Next, we cyclically treated the tumor spheroids with a MEK inhibitor, trametinib, for 6-day periods with a recovery phase in between. This inhibitor was selected because of mutation of colon cancer cells in the MEK/ERK pathway. We used size measurements to evaluate the efficacy of trametinib and predict development of resistance of colon cancer cells during the cyclical treatment and recovery regimen. This size-based analysis closely matched the biochemical analysis of drug responses of spheroids. We performed molecular analysis and showed that resistance to trametinib emerged due to feedback activation of the PI3K/AKT signaling pathway. Therefore, we combined trametinib with a PI3K/AKT inhibitor, dactolisib, and demonstrated that size-based analysis of spheroids reliably allowed quantifying the effect of the combination treatment to prevent drug resistance. This study established that size measurements of spheroids can be used as a straightforward method for quantitative studies of drug responses of tumor spheroids and identifying drug combinations that block resistance.
Collapse
Affiliation(s)
| | - Megha Gupta
- 2 Department of Arts and Sciences, The University of Akron, Akron, Ohio
| | - Madison Plaster
- 1 Department of Biomedical Engineering and The University of Akron, Akron, Ohio
| | - Hossein Tavana
- 1 Department of Biomedical Engineering and The University of Akron, Akron, Ohio
| |
Collapse
|
46
|
Singh S, Tran S, Putman J, Tavana H. Three-dimensional models of breast cancer-fibroblasts interactions. Exp Biol Med (Maywood) 2020; 245:879-888. [PMID: 32276543 DOI: 10.1177/1535370220917366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Tumor stroma plays an important role in progression of cancers to a fatal metastatic disease. Modern treatment strategies are considering targeting tumor stroma to improve outcomes for cancer patients. A current challenge to develop stroma-targeting therapeutics is the lack of preclinical physiologic tumor models. Animal models widely used in cancer research lack human stroma and are not amenable to screening of chemical compounds for cancer drug discovery. In this review, we outline in vitro three-dimensional tumor models that we have developed to study the interactions among cancer cells and stromal cells. We describe development of the tumor models in a modular fashion, from a spheroid model to a sophisticated organotypic model, and discuss the importance of using correct physiologic models to recapitulate tumor-stromal signaling. These biomimetic tumor models will facilitate understanding of tumor-stromal signaling biology and provide a scalable approach for testing and discovery of cancer drugs.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Sydnie Tran
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Justin Putman
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
47
|
Hepato(Geno)Toxicity Assessment of Nanoparticles in a HepG2 Liver Spheroid Model. NANOMATERIALS 2020; 10:nano10030545. [PMID: 32197356 PMCID: PMC7153628 DOI: 10.3390/nano10030545] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022]
Abstract
(1) In compliance with the 3Rs policy to reduce, refine and replace animal experiments, the development of advanced in vitro models is needed for nanotoxicity assessment. Cells cultivated in 3D resemble organ structures better than 2D cultures. This study aims to compare cytotoxic and genotoxic responses induced by titanium dioxide (TiO2), silver (Ag) and zinc oxide (ZnO) nanoparticles (NPs) in 2D monolayer and 3D spheroid cultures of HepG2 human liver cells. (2) NPs were characterized by electron microscopy, dynamic light scattering, laser Doppler anemometry, UV-vis spectroscopy and mass spectrometry. Cytotoxicity was investigated by the alamarBlue assay and confocal microscopy in HepG2 monolayer and spheroid cultures after 24 h of NP exposure. DNA damage (strand breaks and oxidized base lesions) was measured by the comet assay. (3) Ag-NPs were aggregated at 24 h, and a substantial part of the ZnO-NPs was dissolved in culture medium. Ag-NPs induced stronger cytotoxicity in 2D cultures (EC50 3.8 µg/cm2) than in 3D cultures (EC50 > 30 µg/cm2), and ZnO-NPs induced cytotoxicity to a similar extent in both models (EC50 10.1-16.2 µg/cm2). Ag- and ZnO-NPs showed a concentration-dependent genotoxic effect, but the effect was not statistically significant. TiO2-NPs showed no toxicity (EC50 > 75 µg/cm2). (4) This study shows that the HepG2 spheroid model is a promising advanced in vitro model for toxicity assessment of NPs.
Collapse
|
48
|
Zuchowska A, Kasprzak A, Dabrowski B, Kaminska K, Poplawska M, Brzozka Z. Nanoconjugates of graphene oxide derivatives and meso-tetraphenylporphyrin: a new avenue for anticancer photodynamic therapies – Cell-on-a-Chip analysis. NEW J CHEM 2020. [DOI: 10.1039/d0nj04189k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cell-on-a-Chip analysis of nanoconjugates of graphene oxide derivatives and mesotetraphenylporphyrin as a new selective and effective avenue for anticancer photodynamic therapies.
Collapse
Affiliation(s)
- A. Zuchowska
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| | - A. Kasprzak
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| | - B. Dabrowski
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| | - K. Kaminska
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| | - M. Poplawska
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| | - Z. Brzozka
- Chair of Medical Biotechnology
- Warsaw University of Technology
- Faculty of Chemistry
- Warsaw University of Technology
- Poland
| |
Collapse
|
49
|
Plaster M, Singh S, Tavana H. Fibroblasts Promote Proliferation and Matrix Invasion of Breast Cancer Cells in Co‐Culture Models. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Madison Plaster
- Department of Biomedical Engineering The University of Akron Akron OH 44325 USA
| | - Sunil Singh
- Department of Biomedical Engineering The University of Akron Akron OH 44325 USA
| | - Hossein Tavana
- Department of Biomedical Engineering The University of Akron Akron OH 44325 USA
| |
Collapse
|
50
|
Elie BT, Hubbard K, Pechenyy Y, Layek B, Prabha S, Contel M. Preclinical evaluation of an unconventional ruthenium-gold-based chemotherapeutic: RANCE-1, in clear cell renal cell carcinoma. Cancer Med 2019; 8:4304-4314. [PMID: 31192543 PMCID: PMC6675714 DOI: 10.1002/cam4.2322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND There are few effective treatments for patients with advanced clear cell renal cell carcinoma (CCRCC). Recent findings indicate that ruthenium-gold containing compounds exhibit significant antitumor efficacy against CCRCC in vitro affecting cell viability as well as angiogenesis and markers driving those 2 phenomena. However, no in vivo preclinical evaluation of this class of compounds has been reported. METHODS Following the dose-finding pharmacokinetic determination, NOD.CB17-Prkdc SCID/J mice bearing xenograft CCRCC Caki-1 tumors were treated in an intervention trial for 21 days at 10 mg/kg/72h of RANCE-1. At the end of the trial, tumor samples were analyzed for histopathological and changes in protein expression levels were assessed. RESULTS After 21 days of treatment there was no significant change in tumor size in the RANCE-1-treated mice as compared to the starting size (+3.87%) (P = 0.082) while the vehicle treated mice exhibited a significant tumor size increase (+138%) (P < 0.01). There were no signs of pathological complications as a result of treatment. Significant reduction in the expression of VEGF, PDGF, FGF, EGFR, and HGRF, all key to the proliferation of tumor cells and stromal cells serving protumorigenic purposes was observed. CONCLUSIONS The tumor growth inhibition displayed and favorable pathology profile of RANCE-1 makes it a promising candidate for further evaluation toward clinical use for the treatment of advanced CCRCC.
Collapse
Affiliation(s)
- Benelita T. Elie
- Department of ChemistryBrooklyn College, The City University of New YorkBrooklynNew York
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
| | - Karen Hubbard
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Department of BiologyCity College of New York, The City University of New YorkNew YorkNew York
| | - Yuriy Pechenyy
- Department of BiologyCity College of New York, The City University of New YorkNew YorkNew York
| | - Buddhadev Layek
- University of Minnesota College of PharmacyMinneapolisMinnesota
| | - Swayam Prabha
- University of Minnesota College of PharmacyMinneapolisMinnesota
| | - Maria Contel
- Department of ChemistryBrooklyn College, The City University of New YorkBrooklynNew York
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Chemistry PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Biochemistry PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
| |
Collapse
|