51
|
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
|
52
|
Thakuri PS, Gupta M, Joshi R, Singh S, Tavana H. Synergistic Inhibition of Kinase Pathways Overcomes Resistance of Colorectal Cancer Spheroids to Cyclic Targeted Therapies. ACS Pharmacol Transl Sci 2019; 2:275-284. [PMID: 32259061 DOI: 10.1021/acsptsci.9b00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Indexed: 12/11/2022]
Abstract
Cancer cells often adapt to single-agent treatments with chemotherapeutics. Activation of alternative survival pathways is a major mechanism of drug resistance. A potential approach to block this feedback signaling is using combination treatments of a pair of drugs, although toxicity has been a limiting factor. Preclinical tumor models to identify mechanisms of drug resistance and determine low but effective combination doses are critical to effectively suppress tumor growth with reduced toxicity to patients. Using our aqueous two-phase system microtechnology, we developed colorectal tumor spheroids in high-throughput and evaluated resistance of cancer cells to three mitogen-activated protein kinase inhibitors (MAPKi) in long-term cyclic treatments. Our quantitative analysis showed that the efficacy of MAPKi significantly reduced over time, leading to an increase in proliferation of HCT116 colorectal cancer cells and growth of spheroids. We established that resistance was due to feedback activation of PI3K/AKT/mTOR pathway. Using high-throughput, dose-dependent combinations of each MAPKi and a PI3K/mTOR inhibitor, we identified low-dose, synergistic combinations that blocked resistance to MAPKi and effectively suppressed the growth of colorectal tumor spheroids in long-term treatments. Our approach to study drug resistance offers the potential to determine high priority treatments to test in animal models.
Collapse
Affiliation(s)
- Pradip Shahi Thakuri
- 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
| | - Ramila Joshi
- 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
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
53
|
Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G. Integrated cancer tissue engineering models for precision medicine. PLoS One 2019; 14:e0216564. [PMID: 31075118 PMCID: PMC6510431 DOI: 10.1371/journal.pone.0216564] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors are not merely cancerous cells that undergo mindless proliferation. Rather, they are highly organized and interconnected organ systems. Tumor cells reside in complex microenvironments in which they are subjected to a variety of physical and chemical stimuli that influence cell behavior and ultimately the progression and maintenance of the tumor. As cancer bioengineers, it is our responsibility to create physiologic models that enable accurate understanding of the multi-dimensional structure, organization, and complex relationships in diverse tumor microenvironments. Such models can greatly expedite clinical discovery and translation by closely replicating the physiological conditions while maintaining high tunability and control of extrinsic factors. In this review, we discuss the current models that target key aspects of the tumor microenvironment and their role in cancer progression. In order to address sources of experimental variation and model limitations, we also make recommendations for methods to improve overall physiologic reproducibility, experimental repeatability, and rigor within the field. Improvements can be made through an enhanced emphasis on mathematical modeling, standardized in vitro model characterization, transparent reporting of methodologies, and designing experiments with physiological metrics. Taken together these considerations will enhance the relevance of in vitro tumor models, biological understanding, and accelerate treatment exploration ultimately leading to improved clinical outcomes. Moreover, the development of robust, user-friendly models that integrate important stimuli will allow for the in-depth study of tumors as they undergo progression from non-transformed primary cells to metastatic disease and facilitate translation to a wide variety of biological and clinical studies.
Collapse
Affiliation(s)
- Michael E. Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric N. Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Caymen M. Novak
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine S. Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
54
|
Huang Y, Zou J, Badar M, Liu J, Shi W, Wang S, Guo Q, Wang X, Kessel S, Chan LLY, Li P, Liu Y, Qiu J, Zhou C. Longitudinal Morphological and Physiological Monitoring of Three-dimensional Tumor Spheroids Using Optical Coherence Tomography. J Vis Exp 2019. [PMID: 30799861 DOI: 10.3791/59020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Tumor spheroids have been developed as a three-dimensional (3D) cell culture model in cancer research and anti-cancer drug discovery. However, currently, high-throughput imaging modalities utilizing bright field or fluorescence detection, are unable to resolve the overall 3D structure of the tumor spheroid due to limited light penetration, diffusion of fluorescent dyes and depth-resolvability. Recently, our lab demonstrated the use of optical coherence tomography (OCT), a label-free and non-destructive 3D imaging modality, to perform longitudinal characterization of multicellular tumor spheroids in a 96-well plate. OCT was capable of obtaining 3D morphological and physiological information of tumor spheroids growing up to about 600 µm in height. In this article, we demonstrate a high-throughput OCT (HT-OCT) imaging system that scans the whole multi-well plate and obtains 3D OCT data of tumor spheroids automatically. We describe the details of the HT-OCT system and construction guidelines in the protocol. From the 3D OCT data, one can visualize the overall structure of the spheroid with 3D rendered and orthogonal slices, characterize the longitudinal growth curve of the tumor spheroid based on the morphological information of size and volume, and monitor the growth of the dead-cell regions in the tumor spheroid based on optical intrinsic attenuation contrast. We show that HT-OCT can be used as a high-throughput imaging modality for drug screening as well as characterizing biofabricated samples.
Collapse
Affiliation(s)
- Yongyang Huang
- Department of Electrical and Computer Engineering, Lehigh University
| | - Jinyun Zou
- Department of Electrical and Computer Engineering, Lehigh University
| | - Mudabbir Badar
- Department of Electrical and Computer Engineering, Lehigh University
| | - Junchao Liu
- Department of Electrical and Computer Engineering, Lehigh University
| | - Wentao Shi
- Department of Bioengineering, Lehigh University
| | | | - Qiongyu Guo
- Department of Biomedical Engineering, Southern University of Science and Technology
| | - Xiaofang Wang
- Department of Electrical and Computer Engineering, Lehigh University
| | - Sarah Kessel
- Department of Technology R&D, Nexcelom Bioscience LLC
| | | | - Peter Li
- Department of Technology R&D, Nexcelom Bioscience LLC
| | - Yaling Liu
- Department of Mechanical Engineering, Lehigh University; Department of Bioengineering, Lehigh University
| | - Jean Qiu
- Department of Technology R&D, Nexcelom Bioscience LLC
| | - Chao Zhou
- Department of Electrical and Computer Engineering, Lehigh University; Department of Bioengineering, Lehigh University; Center for Photonics and Nanoelectronics, Lehigh University;
| |
Collapse
|
55
|
Shahi Thakuri P, Luker GD, Tavana H. Cyclical Treatment of Colorectal Tumor Spheroids Induces Resistance to MEK Inhibitors. Transl Oncol 2018; 12:404-416. [PMID: 30550927 PMCID: PMC6299152 DOI: 10.1016/j.tranon.2018.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022] Open
Abstract
Adaptive drug resistance is a major obstacle to successful treatment of colorectal cancers. Physiologic tumor models of drug resistance are crucial to understand mechanisms of treatment failure and improve therapy by developing new therapeutics and treatment strategies. Using our aqueous two-phase system microtechnology, we developed colorectal tumor spheroids and periodically treated them with sub-lethal concentrations of three Mitogen Activated Kinase inhibitors (MEKi) used in clinical trials. We used long-term, periodic treatment and recovery of spheroids to mimic cycles of clinical chemotherapy and implemented a growth rate metric to quantitatively assess efficacy of the MEKi during treatment. Our results showed that efficacy of the MEKi significantly reduced with increased treatment cycles. Using a comprehensive molecular analysis, we established that resistance of colorectal tumor spheroids to the MEKi developed through activation of the PI3K/AKT/mTOR pathway. We also showed that other potential feedback mechanisms, such as STAT3 activation or amplified B-RAF, did not account for resistance to the MEKi. We combined each of the three MEKi with a PI3K/mTOR inhibitor and showed that the combination treatments synergistically blocked resistance to the MEKi. Importantly, and unlike the individual inhibitors, we demonstrated that synergistic concentrations of combinations of MEK and PI3K/mTOR inhibitors effectively inhibited growth of colorectal tumor spheroids in long-term treatments. This proof-of-concept study to model treatment-induced drug resistance of cancer cells using 3D cultures offers a unique approach to identify underlying molecular mechanisms and develop effective treatments.
Collapse
Affiliation(s)
- Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48105, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA.
| |
Collapse
|
56
|
Hamilton G, Rath B. Applicability of tumor spheroids for in vitro chemosensitivity assays. Expert Opin Drug Metab Toxicol 2018; 15:15-23. [DOI: 10.1080/17425255.2019.1554055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Gerhard Hamilton
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
57
|
Nunes AS, Barros AS, Costa EC, Moreira AF, Correia IJ. 3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs. Biotechnol Bioeng 2018; 116:206-226. [DOI: 10.1002/bit.26845] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/30/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Ana S. Nunes
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Andreia S. Barros
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Elisabete C. Costa
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - André F. Moreira
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Ilídio J. Correia
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
- Departamento de Engenharia Química; Universidade de Coimbra, (CIEPQF); Coimbra Portugal
| |
Collapse
|
58
|
Singh S, Tavana H. Collagen Partition in Polymeric Aqueous Two-Phase Systems for Tissue Engineering. Front Chem 2018; 6:379. [PMID: 30234101 PMCID: PMC6132203 DOI: 10.3389/fchem.2018.00379] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/03/2018] [Indexed: 12/14/2022] Open
Abstract
Aqueous two-phase systems (ATPS) of polyethylene glycol (PEG) and dextran (DEX) are commonly used to partition proteins. Protein partition in ATPS is a complex phenomenon and depends on factors including molecular weight of polymers, and electrochemical and ionic properties of the phases. We studied the effect of polymer molecular weight on the partition of a natural protein, collagen, in several ATPS formulations made with non-ionic polymers polyethylene glycol (PEG) and dextran (DEX). We found that partitioning of collagen to an aqueous phase significantly increases when the molecular weight of the corresponding phase polymer decreases. Additionally, a large difference between the molecular weight of the phase-forming polymers was necessary to cause a significant uneven collagen distribution between the aqueous phases. We then employed one of the systems to create a three-dimensional breast cancer microtissue by entrapping a spheroid of breast cancer cells within the partitioned collagen. This convenient technique to generate 3D microtissues offers a convenient and promising approach for tissue engineering applications.
Collapse
Affiliation(s)
- Sunil Singh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, United States
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH, United States
| |
Collapse
|
59
|
Miyamoto D, Hara T, Hyakutake A, Nakazawa K. Changes in HepG2 spheroid behavior induced by differences in the gap distance between spheroids in a micropatterned culture system. J Biosci Bioeng 2018; 125:729-735. [DOI: 10.1016/j.jbiosc.2017.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/22/2017] [Accepted: 12/19/2017] [Indexed: 12/13/2022]
|
60
|
Hagemann J, Jacobi C, Gstoettner S, Welz C, Schwenk-Zieger S, Stauber R, Strieth S, Kuenzel J, Baumeister P, Becker S. Therapy Testing in a Spheroid-based 3D Cell Culture Model for Head and Neck Squamous Cell Carcinoma. J Vis Exp 2018. [PMID: 29733308 DOI: 10.3791/57012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Current treatment options for advanced and recurrent head and neck squamous cell carcinoma (HNSCC) enclose radiation and chemo-radiation approaches with or without surgery. While platinum-based chemotherapy regimens currently represent the gold standard in terms of efficacy and are given in the vast majority of cases, new chemotherapy regimens, namely immunotherapy are emerging. However, the response rates and therapy resistance mechanisms for either chemo regimen are hard to predict and remain insufficiently understood. Broad variations of chemo and radiation resistance mechanisms are known to date. This study describes the development of a standardized, high-throughput in vitro assay to assess HNSCC cell line's response to various therapy regimens, and hopefully on primary cells from individual patients as a future tool for personalized tumor therapy. The assay is designed to being integrated into the quality-controlled standard algorithm for HNSCC patients at our tertiary care center; however, this will be subject of future studies. Technical feasibility looks promising for primary cells from tumor biopsies from actual patients. Specimens are then transferred into the laboratory. Biopsies are mechanically separated followed by enzymatic digestion. Cells are then cultured in ultra-low adhesion cell culture vials that promote the reproducible, standardized and spontaneous formation of three-dimensional, spheroid-shaped cell conglomerates. Spheroids are then ready to be exposed to chemo-radiation protocols and immunotherapy protocols as needed. The final cell viability and spheroid size are indicators of therapy susceptibility and therefore could be drawn into consideration in future to assess the patients' likely therapy response. This model could be a valuable, cost-efficient tool towards personalized therapy for head and neck cancer.
Collapse
Affiliation(s)
- Jan Hagemann
- Department of Otorhinolaryngology, Johannes-Gutenberg University Medical Center;
| | - Christian Jacobi
- Department of Otorhinolaryngology, Technical University of Munich Medical Center
| | - Sabine Gstoettner
- Department of Otorhinolaryngology, Ludwig-Maximilian-University Medical Center
| | - Christian Welz
- Department of Otorhinolaryngology, University of Goettingen Medical Center
| | | | - Roland Stauber
- Department of Otorhinolaryngology, Johannes-Gutenberg University Medical Center
| | - Sebastian Strieth
- Department of Otorhinolaryngology, Johannes-Gutenberg University Medical Center
| | - Julian Kuenzel
- Department of Otorhinolaryngology, Johannes-Gutenberg University Medical Center
| | - Philipp Baumeister
- Department of Otorhinolaryngology, Ludwig-Maximilian-University Medical Center
| | - Sven Becker
- Department of Otorhinolaryngology, Johannes-Gutenberg University Medical Center; Department of Otorhinolaryngology, Ludwig-Maximilian-University Medical Center
| |
Collapse
|
61
|
Liu Q, Zhang Z, Liu Y, Cui Z, Zhang T, Li Z, Ma W. Cancer cells growing on perfused 3D collagen model produced higher reactive oxygen species level and were more resistant to cisplatin compared to the 2D model. J Appl Biomater Funct Mater 2018; 16:144-150. [PMID: 29609492 DOI: 10.1177/2280800018764763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Three-dimensional (3D) collagen scaffold models, due to their ability to mimic the tissue and organ structure in vivo, have received increasing interest in drug discovery and toxicity evaluation. METHODS In this study, we developed a perfused 3D model and studied cellular response to cytotoxic drugs in comparison with traditional 2D cell cultures as evaluated by cancer drug cisplatin. RESULTS Cancer cells grown in perfused 3D environments showed increased levels of reactive oxygen species (ROS) production compared to the 2D culture. As determined by growth analysis, cells in the 3D culture, after forming a spheroid, were more resistant to the cancer drug cisplatin compared to that of the 2D cell culture. In addition, 3D culturing cells showed elevated level of ROS, indicating a physiological change or the formation of a microenvironment that resembles tumor cells in vivo. CONCLUSIONS These data revealed that cellular response to drugs for cells growing in 3D environments are dramatically different from that of 2D cultured cells. Thus, the perfused 3D collagen scaffold model we report here might be a potentially very useful tool for drug analysis.
Collapse
Affiliation(s)
- Qingxi Liu
- 1 College of Biotechnology, Tianjin University of Science and Technology, China.,2 Tianjin Weikai Bioeng Ltd., Tianjin, China
| | - Zijiang Zhang
- 1 College of Biotechnology, Tianjin University of Science and Technology, China
| | - Yupeng Liu
- 1 College of Biotechnology, Tianjin University of Science and Technology, China
| | - Zhanfeng Cui
- 3 Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Tongcun Zhang
- 1 College of Biotechnology, Tianjin University of Science and Technology, China
| | - Zhaohui Li
- 3 Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Wenjian Ma
- 1 College of Biotechnology, Tianjin University of Science and Technology, China
| |
Collapse
|
62
|
Thakuri PS, Liu C, Luker GD, Tavana H. Biomaterials-Based Approaches to Tumor Spheroid and Organoid Modeling. Adv Healthc Mater 2018; 7:e1700980. [PMID: 29205942 PMCID: PMC5867257 DOI: 10.1002/adhm.201700980] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/21/2017] [Indexed: 12/22/2022]
Abstract
Evolving understanding of structural and biological complexity of tumors has stimulated development of physiologically relevant tumor models for cancer research and drug discovery. A major motivation for developing new tumor models is to recreate the 3D environment of tumors and context-mediated functional regulation of cancer cells. Such models overcome many limitations of standard monolayer cancer cell cultures. Under defined culture conditions, cancer cells self-assemble into 3D constructs known as spheroids. Additionally, cancer cells may recapitulate steps in embryonic development to self-organize into 3D cultures known as organoids. Importantly, spheroids and organoids reproduce morphology and biologic properties of tumors, providing valuable new tools for research, drug discovery, and precision medicine in cancer. This Progress Report discusses uses of both natural and synthetic biomaterials to culture cancer cells as spheroids or organoids, specifically highlighting studies that demonstrate how these models recapitulate key properties of native tumors. The report concludes with the perspectives on the utility of these models and areas of need for future developments to more closely mimic pathologic events in tumors.
Collapse
Affiliation(s)
- Pradip Shahi Thakuri
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Chun Liu
- Departments of Radiology, Biomedical Engineering and Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gary D Luker
- Departments of Radiology, Biomedical Engineering and Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| |
Collapse
|
63
|
Larsen CJ. [Spheroids: A reference model for in vitro culture of solid tumors?]. Bull Cancer 2017; 105:25-34. [PMID: 29224886 DOI: 10.1016/j.bulcan.2017.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 01/16/2023]
Abstract
The recognition that solid tumors are complex entities composed of the tumor cell mass itself and a stromal micro-environnement providing a variety of cells from the host (fibroblasts, endothelial cells, immune cells) led to recognize that this heterogeneity could not be recapitulated in vitro by conventional bidimensional (2-D) cultures. This justified numerous attempts to develop tridimensional (3-D) cultures that provided better tools for approaching tumor complexity and more convincing drug testing systems. Among various 3-D technologies, tumor spheroids are more likely suited to provide in vitro platforms for apprehending specific aspects of different processes specifically defining each tumor category as well as testing drug delivery systems. This review summarizes current features of multicellular tumor spheroids and their suitability for studying different aspects of cancer cell biology, patient-specific therapies and drug treatment.
Collapse
|
64
|
Ham SL, Thakuri PS, Plaster M, Li J, Luker KE, Luker GD, Tavana H. Three-dimensional tumor model mimics stromal - breast cancer cells signaling. Oncotarget 2017; 9:249-267. [PMID: 29416611 PMCID: PMC5787462 DOI: 10.18632/oncotarget.22922] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/09/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor stroma is a major contributor to the biological aggressiveness of cancer cells. Cancer cells induce activation of normal fibroblasts to carcinoma-associated fibroblasts (CAFs), which promote survival, proliferation, metastasis, and drug resistance of cancer cells. A better understanding of these interactions could lead to new, targeted therapies for cancers with limited treatment options, such as triple negative breast cancer (TNBC). To overcome limitations of standard monolayer cell cultures and xenograft models that lack tumor complexity and/or human stroma, we have developed a high throughput tumor spheroid technology utilizing a polymeric aqueous two-phase system to conveniently model interactions of CAFs and TNBC cells and quantify effects on signaling and drug resistance of cancer cells. We focused on signaling by chemokine CXCL12, a hallmark molecule secreted by CAFs, and receptor CXCR4, a driver of tumor progression and metastasis in TNBC. Using three-dimensional stromal-TNBC cells cultures, we demonstrate that CXCL12 – CXCR4 signaling significantly increases growth of TNBC cells and drug resistance through activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways. Despite resistance to standard chemotherapy, upregulation of MAPK and PI3K signaling sensitizes TNBC cells in co-culture spheroids to specific inhibitors of these kinase pathways. Furthermore, disrupting CXCL12 – CXCR4 signaling diminishes drug resistance of TNBC cells in co-culture spheroid models. This work illustrates the capability to identify mechanisms of drug resistance and overcome them using our engineered model of tumor-stromal interactions.
Collapse
Affiliation(s)
- Stephanie Lemmo Ham
- 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
| | - Madison Plaster
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Jun Li
- Department of Mathematical Sciences, Kent State University, Kent, OH 44242, USA
| | - Kathryn E Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| |
Collapse
|
65
|
Costa EC, de Melo-Diogo D, Moreira AF, Carvalho MP, Correia IJ. Spheroids Formation on Non-Adhesive Surfaces by Liquid Overlay Technique: Considerations and Practical Approaches. Biotechnol J 2017; 13. [DOI: 10.1002/biot.201700417] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/03/2017] [Accepted: 10/16/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Elisabete C. Costa
- CICS-UBI − Health Sciences Research Centre; Universidade da Beira Interior; 6200-506 Covilhã Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI − Health Sciences Research Centre; Universidade da Beira Interior; 6200-506 Covilhã Portugal
| | - André F. Moreira
- CICS-UBI − Health Sciences Research Centre; Universidade da Beira Interior; 6200-506 Covilhã Portugal
| | - Marco P. Carvalho
- CICS-UBI − Health Sciences Research Centre; Universidade da Beira Interior; 6200-506 Covilhã Portugal
| | - Ilídio J. Correia
- CICS-UBI − Health Sciences Research Centre; Universidade da Beira Interior; 6200-506 Covilhã Portugal
| |
Collapse
|
66
|
Huang Y, Wang S, Guo Q, Kessel S, Rubinoff I, Chan LLY, Li P, Liu Y, Qiu J, Zhou C. Optical Coherence Tomography Detects Necrotic Regions and Volumetrically Quantifies Multicellular Tumor Spheroids. Cancer Res 2017; 77:6011-6020. [PMID: 28904062 DOI: 10.1158/0008-5472.can-17-0821] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/24/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) tumor spheroid models have gained increased recognition as important tools in cancer research and anticancer drug development. However, currently available imaging approaches used in high-throughput screening drug discovery platforms, for example, bright-field, phase contrast, and fluorescence microscopies, are unable to resolve 3D structures deep inside (>50 μm) tumor spheroids. In this study, we established a label-free, noninvasive optical coherence tomography (OCT) imaging platform to characterize 3D morphologic and physiologic information of multicellular tumor spheroids (MCTS) growing from approximately 250 to 600 μm in height over 21 days. In particular, tumor spheroids of two cell lines, glioblastoma (U-87MG) and colorectal carcinoma (HCT116), exhibited distinctive evolutions in their geometric shapes at late growth stages. Volumes of MCTS were accurately quantified using a voxel-based approach without presumptions of their geometries. In contrast, conventional diameter-based volume calculations assuming perfect spherical shape resulted in large quantification errors. Furthermore, we successfully detected necrotic regions within these tumor spheroids based on increased intrinsic optical attenuation, suggesting a promising alternative of label-free viability tests in tumor spheroids. Therefore, OCT can serve as a promising imaging modality to characterize morphologic and physiologic features of MCTS, showing great potential for high-throughput drug screening. Cancer Res; 77(21); 6011-20. ©2017 AACR.
Collapse
Affiliation(s)
- Yongyang Huang
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Shunqiang Wang
- Department of Mechanical Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Qiongyu Guo
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Sarah Kessel
- Department of Technology R&D, Nexcelom Bioscience LLC, Lawrence, Massachusetts
| | - Ian Rubinoff
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania
| | - Leo Li-Ying Chan
- Department of Technology R&D, Nexcelom Bioscience LLC, Lawrence, Massachusetts
| | - Peter Li
- Department of Technology R&D, Nexcelom Bioscience LLC, Lawrence, Massachusetts
| | - Yaling Liu
- Department of Mechanical Engineering, Lehigh University, Bethlehem, Pennsylvania.,Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania
| | - Jean Qiu
- Department of Technology R&D, Nexcelom Bioscience LLC, Lawrence, Massachusetts
| | - Chao Zhou
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania. .,Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania.,Center for Photonics and Nanoelectronics, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
67
|
Abstract
The development of 3D cell cultures into self-organizing organ-like structures named organoids provides a model that better reflects in vivo organ physiology and their functional properties. Organoids have been established from several organs, such as the intestine, prostate, brain, liver, kidney and pancreas. With recent advances in high-throughput and -omics profiling technologies, it is now possible to study the mechanisms of cellular organisation at the systems level. It is therefore not surprising that these methods are now used to characterize organoids at the transcriptomic, proteomic, chromatin state and transcription factor DNA-binding levels. These approaches can therefore provide a wealth of information regarding both the mechanisms involved in different diseases, and those involved in cell responses to different conditions, in a more in vivo setting. The authors provide an overview of the potential applications of quantitative mass spectrometry with organoid culture, and how the use of large-scale proteome measurements is emerging in different organoid systems.
Collapse
Affiliation(s)
- Alexis Gonneaud
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Claude Asselin
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Boudreau
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | |
Collapse
|
68
|
Riffle S, Hegde RS. Modeling tumor cell adaptations to hypoxia in multicellular tumor spheroids. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:102. [PMID: 28774341 PMCID: PMC5543535 DOI: 10.1186/s13046-017-0570-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022]
Abstract
Under hypoxic conditions, tumor cells undergo a series of adaptations that promote evolution of a more aggressive tumor phenotype including the activation of DNA damage repair proteins, altered metabolism, and decreased proliferation. Together these changes mitigate the negative impact of oxygen deprivation and allow preservation of genomic integrity and proliferative capacity, thus contributing to tumor growth and metastasis. As a result the presence of a hypoxic microenvironment is considered a negative clinical feature of many solid tumors. Hypoxic niches in tumors also represent a therapeutically privileged environment in which chemo- and radiation therapy is less effective. Although the negative impact of tumor hypoxia has been well established, the precise effect of oxygen deprivation on tumor cell behavior, and the molecular signals that allow a tumor cell to survive in vivo are poorly understood. Multicellular tumor spheroids (MCTS) have been used as an in vitro model for the avascular tumor niche, capable of more accurately recreating tumor genomic profiles and predicting therapeutic response. However, relatively few studies have used MCTS to study the molecular mechanisms driving tumor cell adaptations within the hypoxic tumor environment. Here we will review what is known about cell proliferation, DNA damage repair, and metabolic pathways as modeled in MCTS in comparison to observations made in solid tumors. A more precise definition of the cell populations present within 3D tumor models in vitro could better inform our understanding of the heterogeneity within tumors as well as provide a more representative platform for the testing of therapeutic strategies.
Collapse
Affiliation(s)
- Stephen Riffle
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Rashmi S Hegde
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.
| |
Collapse
|
69
|
Wei Y, Wang Y, Xia D, Guo S, Wang F, Zhang X, Gan Y. Thermosensitive Liposomal Codelivery of HSA-Paclitaxel and HSA-Ellagic Acid Complexes for Enhanced Drug Perfusion and Efficacy Against Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25138-25151. [PMID: 28696100 DOI: 10.1021/acsami.7b07132] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Fibrotic stroma and tumor-promoting pancreatic stellate cells (PSCs), critical characters in the pancreatic ductal adenocarcinoma (PDA) microenvironment, promote a tumor-facilitating environment that simultaneously prevents drug penetration into tumor foci and stimulates tumor growth. Nab-PTX, a human serum albumin (HSA) nanoparticle of paclitaxel (PTX), indicates enhanced matrix penetration in PDA probably due to its small size in vivo and high affinity of HSA with secreted protein acidic and rich in cysteine (SPARC), overexpressed in the PDA stroma. However, this HSA nanoparticle shows poor drug blood retention because of its weak colloidal stability in vivo, thus resulting in insufficient drug accumulation within tumor. Encapsulating HSA nanoparticles into the internal aqueous phase of ordinary liposomes improves their blood retention and the following tumor accumulation, but the large 200 nm size and shielding of HSA in the interior might make it difficult for this hybrid nanomedicine to penetrate the fibrotic PDA matrix and promote bioavailability of the payload. In our current work, we prepared ∼9 nm HSA complexes with an antitumor drug (PTX) and an anti-PSC drug (ellagic acid, EA), and these two HSA-drug complexes were further coencapsulated into thermosensitive liposomes (TSLs). This nanomedicine was named TSL/HSA-PE. The use of TSL/HSA-PE could improve drug blood retention, and upon reaching locally heated tumors, these TSLs can rapidly release their payloads (HSA-drug complexes) to facilitate their further tumor accumulation and matrix penetration. With superior tumor accumulation, impressive matrix penetration, and simultaneous action upon tumor cells and PSCs to disrupt PSCs-PDA interaction, TSL/HSA-PE treatment combined with heat exhibited strong tumor growth inhibition and apoptosis in vivo.
Collapse
Affiliation(s)
- Yan Wei
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yuxi Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Nano Science and Technology Institute, University of Science and Technology of China , 166 Renai Road, Suzhou, Jiangsu 215123, China
| | - Dengning Xia
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Shiyan Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Feng Wang
- Shanghai Institute of Pharmaceutical Industry , 285 Gebaini Road, Shanghai 201203, China
| | - Xinxin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| |
Collapse
|
70
|
Advanced biomaterials and microengineering technologies to recapitulate the stepwise process of cancer metastasis. Biomaterials 2017; 133:176-207. [DOI: 10.1016/j.biomaterials.2017.04.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/04/2017] [Accepted: 04/12/2017] [Indexed: 02/08/2023]
|
71
|
Ishiguro T, Ohata H, Sato A, Yamawaki K, Enomoto T, Okamoto K. Tumor-derived spheroids: Relevance to cancer stem cells and clinical applications. Cancer Sci 2017; 108:283-289. [PMID: 28064442 PMCID: PMC5378268 DOI: 10.1111/cas.13155] [Citation(s) in RCA: 344] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/13/2016] [Accepted: 12/28/2016] [Indexed: 02/06/2023] Open
Abstract
Recently, many types of in vitro 3‐D culture systems have been developed to recapitulate the in vivo growth conditions of cancer. The cancer 3‐D culture methods aim to preserve the biological characteristics of original tumors better than conventional 2‐D monolayer cultures, and include tumor‐derived organoids, tumor‐derived spheroids, organotypic multicellular spheroids, and multicellular tumor spheroids. The 3‐D culture methods differ in terms of cancer cell sources, protocols for cell handling, and the required time intervals. Tumor‐derived spheroids are unique because they are purposed for the enrichment of cancer stem cells (CSCs) or cells with stem cell‐related characteristics. These spheroids are grown as floating spheres and have been used as surrogate systems to evaluate the CSC‐related characteristics of solid tumors in vitro. Because eradication of CSCs is likely to be of clinical importance due to their association with the malignant nature of cancer cells, such as tumorigenicity or chemoresistance, the investigation of tumor‐derived spheroids may provide invaluable clues to fight against cancer. Spheroid cultures have been established from cancers including glioma, breast, colon, ovary, and prostate cancers, and their biological and biochemical characteristics have been investigated by many research groups. In addition to the investigation of CSCs, tumor‐derived spheroids may prove to be instrumental for a high‐throughput screening platform or for the cultivation of CSC‐related tumor cells found in the circulation or body fluids.
Collapse
Affiliation(s)
- Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Medical School, Niigata, Japan
| | - Hirokazu Ohata
- Division of Cancer Differentiation, National Cancer Center Research Institute, Tokyo, Japan
| | - Ai Sato
- Division of Cancer Differentiation, National Cancer Center Research Institute, Tokyo, Japan
| | - Kaoru Yamawaki
- Department of Obstetrics and Gynecology, Niigata University Medical School, Niigata, Japan.,Division of Cancer Differentiation, National Cancer Center Research Institute, Tokyo, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Medical School, Niigata, Japan
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
72
|
Shahi Thakuri P, Tavana H. Single and Combination Drug Screening with Aqueous Biphasic Tumor Spheroids. SLAS DISCOVERY 2017; 22:507-515. [PMID: 28324660 DOI: 10.1177/2472555217698817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spheroids of cancer cells represent a physiologic model of solid tumors for cancer drug screening. Despite this known benefit, difficulties with generating large quantities of uniformly sized spheroids in standard plates, individually addressing spheroids with drug compounds, and quantitatively analyzing responses of cancer cells have hindered the use of spheroids in high-throughput screening applications. Recently, we addressed this challenge by using an aqueous two-phase system technology to generate a spheroid within an aqueous drop immersed in a second, immiscible aqueous phase. Integrating this approach with robotics resulted in convenient formation, maintenance, and drug treatment of spheroids. Here, we demonstrate the feasibility of high-throughput compound screening against colon cancer spheroids using 25 anticancer compounds. Using a strictly standardized mean difference and based on a preliminary testing with each compound, we select effective compounds for further dose-response testing. Finally, we use molecular inhibitors to target upregulated protein kinases and use them for drug combination studies against spheroids. We quantitatively analyze the combination treatment results using statistical metrics to identify synergy between pairs of inhibitors in compromising viability of colon cancer cells. This study demonstrates the utility of our spheroid culture technology for identification of effective drug compounds, dose-response analysis, and combination drug treatments.
Collapse
Affiliation(s)
| | - Hossein Tavana
- 1 Department of Biomedical Engineering, The University of Akron, Akron, OH, USA
| |
Collapse
|
73
|
Ham SL, Joshi R, Luker GD, Tavana H. Engineered Breast Cancer Cell Spheroids Reproduce Biologic Properties of Solid Tumors. Adv Healthc Mater 2016; 5:2788-2798. [PMID: 27603912 PMCID: PMC5142748 DOI: 10.1002/adhm.201600644] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/03/2016] [Indexed: 01/11/2023]
Abstract
Solid tumors develop as 3D tissue constructs. As tumors grow larger, spatial gradients of nutrients and oxygen and inadequate diffusive supply to cells distant from vasculature develops. Hypoxia initiates signaling and transcriptional alterations to promote survival of cancer cells and generation of cancer stem cells (CSCs) that have self-renewal and tumor-initiation capabilities. Both hypoxia and CSCs are associated with resistance to therapies and tumor relapse. This study demonstrates that 3D cancer cell models, known as tumor spheroids, generated with a polymeric aqueous two-phase system (ATPS) technology capture these important biological processes. Similar to solid tumors, spheroids of triple negative breast cancer cells deposit major extracellular matrix proteins. The molecular analysis establishes presence of hypoxic cells in the core region and expression of CSC gene and protein markers including CD24, CD133, and Nanog. Importantly, these spheroids resist treatment with chemotherapy drugs. A combination treatment approach using a hypoxia-activated prodrug, TH-302, and a chemotherapy drug, doxorubicin, successfully targets drug resistant spheroids. This study demonstrates that ATPS spheroids recapitulate important biological and functional properties of solid tumors and provide a unique model for studies in cancer research.
Collapse
Affiliation(s)
- Stephanie L. Ham
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States
| | - Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States
| | - Gary D. Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, United States
| |
Collapse
|
74
|
Yue X, Lukowski JK, Weaver EM, Skube SB, Hummon AB. Quantitative Proteomic and Phosphoproteomic Comparison of 2D and 3D Colon Cancer Cell Culture Models. J Proteome Res 2016; 15:4265-4276. [PMID: 27696853 DOI: 10.1021/acs.jproteome.6b00342] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell cultures are widely used model systems. Some immortalized cell lines can be grown in either two-dimensional (2D) adherent monolayers or in three-dimensional (3D) multicellular aggregates, or spheroids. Here, the quantitative proteome and phosphoproteome of colon carcinoma HT29 cells cultures in 2D monolayers and 3D spheroids were compared with a stable isotope labeling of amino acids (SILAC) labeling strategy. Two biological replicates from each sample were examined, and notable differences in both the proteome and the phosphoproteome were determined by nanoliquid chromatography tandem mass spectrometry (LC-MS/MS) to assess how growth configuration affects molecular expression. A total of 5867 protein groups, including 2523 phosphoprotein groups and 8733 phosphopeptides were identified in the samples. The Gene Ontology analysis revealed enriched GO terms in the 3D samples for RNA binding, nucleic acid binding, enzyme binding, cytoskeletal protein binding, and histone binding for their molecular functions (MF) and in the process of cell cycle, cytoskeleton organization, and DNA metabolic process for the biological process (BP). The KEGG pathway analysis indicated that 3D cultures are enriched for oxidative phosphorylation pathways, metabolic pathways, peroxisome pathways, and biosynthesis of amino acids. In contrast, analysis of the phosphoproteomes indicated that 3D cultures have decreased phosphorylation correlating with slower growth rates and lower cell-to-extracellular matrix interactions. In sum, these results provide quantitative assessments of the effects on the proteome and phosphoproteome of culturing cells in 2D versus 3D cell culture configurations.
Collapse
Affiliation(s)
- Xiaoshan Yue
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame , 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Jessica K Lukowski
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame , 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Eric M Weaver
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame , 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Susan B Skube
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame , 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame , 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
75
|
Shahi Thakuri P, Ham SL, Luker GD, Tavana H. Multiparametric Analysis of Oncology Drug Screening with Aqueous Two-Phase Tumor Spheroids. Mol Pharm 2016; 13:3724-3735. [DOI: 10.1021/acs.molpharmaceut.6b00527] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Pradip Shahi Thakuri
- Department
of Biomedical Engineering, The University of Akron, Akron, Ohio 44325 United States
| | - Stephanie L. Ham
- Department
of Biomedical Engineering, The University of Akron, Akron, Ohio 44325 United States
| | - Gary D. Luker
- Departments
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
|