1
|
Lenart NA, Rao SS. Cell-cell interactions mediating primary and metastatic breast cancer dormancy. Cancer Metastasis Rev 2024; 44:6. [PMID: 39585533 DOI: 10.1007/s10555-024-10223-5] [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: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Breast cancer remains one of the leading causes of death in women around the world. A majority of deaths from breast cancer occur due to cancer cells colonizing distant organ sites. When colonizing these distant organ sites, breast cancer cells have been known to enter into a state of dormancy for extended periods of time. However, the mechanisms that promote dormancy as well as dormant-to-proliferative switch are not fully understood. The tumor microenvironment plays a key role in mediating cancer cell phenotype including regulation of the dormant state. In this review, we highlight cell-cell interactions in the tumor microenvironment mediating breast cancer dormancy at the primary and metastatic sites. Specifically, we describe how immune cells from the lymphoid lineage, tumor-associated myeloid lineage cells, and stromal cells of non-hematopoietic origin as well as tissue resident stromal cells impact dormancy vs. proliferation in breast cancer cells as well as the associated mechanisms. In addition, we highlight the importance of developing model systems and the associated considerations that will be critical in unraveling the mechanisms that promote primary and metastatic breast cancer dormancy mediated via cell-cell interactions.
Collapse
Affiliation(s)
- Nicholas A Lenart
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA.
| |
Collapse
|
2
|
Rafsanjani Nejad P, Lamichhane A, Guragain P, Luker G, Tavana H. A gravity-driven tissue chip to study the efficacy and toxicity of cancer therapeutics. LAB ON A CHIP 2024; 24:5251-5263. [PMID: 39485368 PMCID: PMC11529822 DOI: 10.1039/d4lc00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Tissue chip and organs-on-chip technologies have emerged as promising tools in preclinical studies. In oncology, this is driven by the high failure rates of candidate drugs in clinical trials mainly due to inadequate efficacy or intolerable toxicity and the need for better predictive preclinical models than those traditionally used. However, the intricate design, fabrication, operation, and limited compatibility with automation limit the utility of tissue chips. To tackle these issues, we designed a novel 32-unit tissue chip in the format of standard 96-well plates to streamline automation, fabricated it using 3D printing, and leveraged gravity-driven flow to bypass the need for external flow devices. Each unit includes three interconnected tissue compartments that model liver, tumor, and bone marrow stroma. The focus on liver and bone marrow stroma was due to their respective roles in drug metabolism and disturbances to the bone marrow niche from off-target toxicity of chemotherapies. We analyzed flow patterns, mixing, and oxygen transport among and within the compartments through finite element simulations and demonstrated the utility of the tissue chip to study the efficacy of commonly-used cytotoxic cancer drugs against tumor cells and their toxicity toward liver and bone marrow cells. The ability to simultaneously study drug efficacy and toxicity in high throughput can help select promising therapeutics in early stages of drug discovery in preclinical studies.
Collapse
Affiliation(s)
| | - Astha Lamichhane
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| | - Prasiddha Guragain
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| | - Gary Luker
- Departments of Radiology, Microbiology and Immunology, Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Hossein Tavana
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
| |
Collapse
|
3
|
Sinha S, Callow BW, Farfel AP, Roy S, Chen S, Rajendran S, Buschhaus JM, Espinoza CR, Luker KE, Ghosh P, Luker GD. Breast Cancers That Disseminate to Bone Marrow Acquire Aggressive Phenotypes through CX43-related Tumor-Stroma Tunnels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.18.533175. [PMID: 36993616 PMCID: PMC10055300 DOI: 10.1101/2023.03.18.533175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Estrogen receptor-positive (ER+) breast cancer commonly disseminates to bone marrow, where interactions with mesenchymal stromal cells (MSCs) shape disease trajectory. We modeled these interactions with tumor-MSC co-cultures and used an integrated transcriptome-proteome-network-analyses workflow to identify a comprehensive catalog of contact-induced changes. Conditioned media from MSCs failed to recapitulate genes and proteins, some borrowed and others tumor-intrinsic, induced in cancer cells by direct contact. Protein-protein interaction networks revealed the rich connectome between 'borrowed' and 'intrinsic' components. Bioinformatics prioritized one of the 'borrowed' components, CCDC88A /GIV, a multi-modular metastasis-related protein that has recently been implicated in driving a hallmark of cancer, growth signaling autonomy. MSCs transferred GIV protein to ER+ breast cancer cells (that lack GIV) through tunnelling nanotubes via connexin (Cx)43-facilitated intercellular transport. Reinstating GIV alone in GIV-negative breast cancer cells reproduced ∼20% of both the 'borrowed' and the 'intrinsic' gene induction patterns from contact co-cultures; conferred resistance to anti-estrogen drugs; and enhanced tumor dissemination. Findings provide a multiomic insight into MSC→tumor cell intercellular transport and validate how transport of one such candidate, GIV, from the haves (MSCs) to have-nots (ER+ breast cancer) orchestrates aggressive disease states.
Collapse
|
4
|
Torres Chavez AG, McKenna MK, Balasubramanian K, Riffle L, Patel NL, Kalen JD, St. Croix B, Leen AM, Bajgain P. A dual-luciferase bioluminescence system for the assessment of cellular therapies. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200763. [PMID: 38596291 PMCID: PMC10869576 DOI: 10.1016/j.omton.2024.200763] [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: 09/20/2023] [Revised: 12/17/2023] [Accepted: 01/05/2024] [Indexed: 04/11/2024]
Abstract
Bioluminescence imaging is a well-established platform for evaluating engineered cell therapies in preclinical studies. However, despite the discovery of new luciferases and substrates, optimal combinations to simultaneously monitor two cell populations remain limited. This makes the functional assessment of cellular therapies cumbersome and expensive, especially in preclinical in vivo models. In this study, we explored the potential of using a green bioluminescence-emitting click beetle luciferase, CBG99, and a red bioluminescence-emitting firefly luciferase mutant, Akaluc, together to simultaneously monitor two cell populations. Using various chimeric antigen receptor T cells and tumor pairings, we demonstrate that these luciferases are suitable for real-time tracking of two cell types using 2D and 3D cultures in vitro and experimental models in vivo. Our data show the broad compatibility of this dual-luciferase (duo-luc) system with multiple bioluminescence detection equipment ranging from benchtop spectrophotometers to live animal imaging systems. Although this study focused on investigating complex CAR T cells and tumor cell interactions, this duo-luc system has potential utility for the simultaneous monitoring of any two cellular components-for example, to unravel the impact of a specific genetic variant on clonal dominance in a mixed population of tumor cells.
Collapse
Affiliation(s)
| | - Mary K. McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Lisa Riffle
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD 21702, USA
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
5
|
Sinha S, Farfel A, Luker KE, Parker BA, Yeung KT, Luker GD, Ghosh P. Growth signaling autonomy in circulating tumor cells aids metastatic seeding. PNAS NEXUS 2024; 3:pgae014. [PMID: 38312224 PMCID: PMC10833458 DOI: 10.1093/pnasnexus/pgae014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024]
Abstract
Self-sufficiency (autonomy) in growth signaling, the earliest recognized hallmark of cancer, is fueled by the tumor cell's ability to "secrete-and-sense" growth factors (GFs); this translates into cell survival and proliferation that is self-sustained by autocrine/paracrine secretion. A Golgi-localized circuitry comprised of two GTPase switches has recently been implicated in the orchestration of growth signaling autonomy. Using breast cancer cells that are either endowed or impaired (by gene editing) in their ability to assemble the circuitry for growth signaling autonomy, here we define the transcriptome, proteome, and phenome of such an autonomous state, and unravel its role during cancer progression. We show that autonomy is associated with enhanced molecular programs for stemness, proliferation, and epithelial-mesenchymal plasticity. Autonomy is both necessary and sufficient for anchorage-independent GF-restricted proliferation and resistance to anticancer drugs and is required for metastatic progression. Transcriptomic and proteomic studies show that autonomy is associated, with a surprising degree of specificity, with self-sustained epidermal growth factor receptor (EGFR)/ErbB signaling. Derivation of a gene expression signature for autonomy revealed that growth signaling autonomy is uniquely induced in circulating tumor cells (CTCs), the harshest phase in the life of tumor cells when it is deprived of biologically available epidermal growth factor (EGF). We also show that autonomy in CTCs tracks therapeutic response and prognosticates outcome. These data support a role for growth signaling autonomy in multiple processes essential for the blood-borne dissemination of human breast cancer.
Collapse
Affiliation(s)
- Saptarshi Sinha
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alex Farfel
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Kathryn E Luker
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Barbara A Parker
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kay T Yeung
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Gary D Luker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109-2200, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109-2200, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Veterans Affairs Medical Center, 3350 La Jolla Village Drive, San Diego, CA 92161, USA
| |
Collapse
|
6
|
Humphries BA, Zhang A, Buschhaus JM, Bevoor A, Farfel A, Rajendran S, Cutter AC, Luker GD. Enhanced mitochondrial fission inhibits triple-negative breast cancer cell migration through an ROS-dependent mechanism. iScience 2023; 26:106788. [PMID: 37235049 PMCID: PMC10206500 DOI: 10.1016/j.isci.2023.106788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/27/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria produce reactive oxygen species (ROS), which function in signal transduction. Mitochondrial dynamics, encompassing morphological shifts between fission and fusion, can directly impact ROS levels in cancer cells. In this study, we identified an ROS-dependent mechanism for how enhanced mitochondrial fission inhibits triple negative breast cancer (TNBC) cell migration. We found that enforcing mitochondrial fission in TNBC resulted in an increase in intracellular ROS levels and reduced cell migration and the formation of actin-rich migratory structures. Consistent with mitochondrial fission, increasing ROS levels in cells inhibited cell migration. Conversely, reducing ROS levels with either a global or mitochondrially targeted scavenger overcame the inhibitory effects of mitochondrial fission. Mechanistically, we found that the ROS sensitive SHP-1/2 phosphatases partially regulate inhibitory effects of mitochondrial fission on TNBC migration. Overall, our work reveals the inhibitory effects of ROS in TNBC and supports mitochondrial dynamics as a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Brock A. Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anne Zhang
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Johanna M. Buschhaus
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Avinash Bevoor
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alex Farfel
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shrila Rajendran
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alyssa C. Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gary D. Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
The CXCL12/CXCR4/ACKR3 Signaling Axis Regulates PKM2 and Glycolysis. Cells 2022; 11:cells11111775. [PMID: 35681470 PMCID: PMC9179862 DOI: 10.3390/cells11111775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
In response to CXCL12, CXCR4 and ACKR3 both recruit β-arrestin 2, regulating the assembly of interacting proteins that drive signaling and contribute to the functions of both receptors in cancer and multiple other diseases. A prior proteomics study revealed that β-arrestin 2 scaffolds pyruvate kinase M2 (PKM2), an enzyme implicated in shifting cells to glycolytic metabolism and poor prognosis in cancer. We hypothesized that CXCL12 signaling regulates PKM2 protein interactions, oligomerization, and glucose metabolism. We used luciferase complementation in cell-based assays and a tumor xenograft model of breast cancer in NSG mice to quantify how CXCR4 and ACKR3 change protein interactions in the β-arrestin-ERK-PKM2 pathway. We also used mass spectrometry to analyze the effects of CXCL12 on glucose metabolism. CXCL12 signaling through CXCR4 and ACKR3 stimulated protein interactions among β-arrestin 2, PKM2, ERK2, and each receptor, leading to the dissociation of PKM2 from β-arrestin 2. The activation of both receptors reduced the oligomerization of PKM2, reflecting a shift from tetramers to dimers or monomers with low enzymatic activity. Mass spectrometry with isotopically labeled glucose showed that CXCL12 signaling increased intermediate metabolites in glycolysis and the pentose phosphate pathway, with ACKR3 mediating greater effects. These data establish how CXCL12 signaling regulates PKM2 and reprograms cellular metabolism.
Collapse
|
8
|
Liu C, Li M, Dong ZX, Jiang D, Li X, Lin S, Chen D, Zou X, Zhang XD, Luker GD. Heterogeneous microenvironmental stiffness regulates pro-metastatic functions of breast cancer cells. Acta Biomater 2021; 131:326-340. [PMID: 34246802 PMCID: PMC8784164 DOI: 10.1016/j.actbio.2021.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022]
Abstract
Besides molecular and phenotypic variations observed in cancer cells, intratumoral heterogeneity also occurs in the tumor microenvironment. Correlative stiffness maps of different intratumor locations in breast tumor biopsies show that stiffness increases from core to periphery. However, how different local ECM stiffness regulates key functions of cancer cells in tumor progression remains unclear. Although increased tissue stiffness is an established driver of breast cancer progression, conclusions from 2D cultures do not correspond with newer data from cancer cells in 3D environments. Many past studies of breast cancer in 3D culture fail to recapitulate the stiffness of a real breast tumor or the various local stiffnesses present in a tumor microenvironment. In this study, we developed a series of collagen/alginate hybrid hydrogels with adjustable stiffness to match the core, middle, and peripheral zones of a breast tumor. We used this hydrogel system to investigate effects of different local stiffness on morphology, proliferation, and migration of breast cancer cells. RNA sequencing of cells in hydrogels with different stiffness revealed changes in multiple cellular processes underlying cancer progression, including angiogenesis and metabolism. We discovered that tumor cells in a soft environment enriched YAP1 and AP1 signaling related genes, whereas tumor cells in a stiff environment became more pro-angiogenic by upregulating fibronectin 1 (FN1) and matrix metalloproteinase 9 (MMP9) expression. This systematic study defines how the range of environmental stiffnesses present in a breast tumor regulates cancer cells, providing new insights into tumorigenesis and disease progression at the tumor-stroma interface. STATEMENT OF SIGNIFICANCE: Applied a well-defined hybrid hydrogel system to mimic the tumor microenvironment with heterogeneous local stiffness. Breast cancer cells tended to proliferate in soft core environment while migrate in stiff peripheral environment. Breast cancer cells shift from glycolysis to OXPHOS and fatty acid metabolism responding to stiff matrix microenvironment. The transcriptomic profile of breast cancer cells altered due to microenvironmental stiffness changes.
Collapse
Affiliation(s)
- Chun Liu
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China.
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhao-Xia Dong
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Dong Jiang
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xiaojing Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Demeng Chen
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuenong Zou
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xing-Ding Zhang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
9
|
Bushnell GG, Deshmukh AP, den Hollander P, Luo M, Soundararajan R, Jia D, Levine H, Mani SA, Wicha MS. Breast cancer dormancy: need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer 2021; 7:66. [PMID: 34050189 PMCID: PMC8163741 DOI: 10.1038/s41523-021-00269-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/08/2021] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in the USA. Although advances in treatment over the past several decades have significantly improved the outlook for this disease, most women who are diagnosed with estrogen receptor positive disease remain at risk of metastatic relapse for the remainder of their life. The cellular source of late relapse in these patients is thought to be disseminated tumor cells that reactivate after a long period of dormancy. The biology of these dormant cells and their natural history over a patient's lifetime is largely unclear. We posit that research on tumor dormancy has been significantly limited by the lack of clinically relevant models. This review will discuss existing dormancy models, gaps in biological understanding, and propose criteria for future models to enhance their clinical relevance.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Abhijeet P Deshmukh
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Luo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics and Departments of Physics and Bioengineering, Northeastern University, Boston, MA, USA.
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Falgàs A, Pallarès V, Unzueta U, Núñez Y, Sierra J, Gallardo A, Alba-Castellón L, Mangues MA, Álamo P, Villaverde A, Vázquez E, Mangues R, Casanova I. Specific Cytotoxic Effect of an Auristatin Nanoconjugate Towards CXCR4 + Diffuse Large B-Cell Lymphoma Cells. Int J Nanomedicine 2021; 16:1869-1888. [PMID: 33716502 PMCID: PMC7944372 DOI: 10.2147/ijn.s289733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Around 40-50% of diffuse large-B cell lymphoma (DLBCL) patients suffer from refractory disease or relapse after R-CHOP first-line treatment. Many ongoing clinical trials for DLBCL patients involve microtubule targeting agents (MTAs), however, their anticancer activity is limited by severe side effects. Therefore, we chose to improve the therapeutic window of the MTA monomethyl auristatin E developing a nanoconjugate, T22-AUR, that selectively targets the CXCR4 receptor, which is overexpressed in many DLBCL cells (CXCR4+) and associated with poor prognosis. METHODS The T22-AUR specificity towards CXCR4 receptor was performed by flow cytometry in different DLBCL cell lines and running biodistribution assays in a subcutaneous mouse model bearing CXCR4+ DLBCL cells. Moreover, we determined T22-AUR cytotoxicity using cell viability assays, cell cycle analysis, DAPI staining and immunohistochemistry. Finally, the T22-AUR antineoplastic effect was evaluated in vivo in an extranodal CXCR4+ DLBCL mouse model whereas the toxicity analysis was assessed by histopathology in non-infiltrated mouse organs and by in vitro cytotoxic assays in human PBMCs. RESULTS We demonstrate that the T22-AUR nanoconjugate displays CXCR4-dependent targeting and internalization in CXCR4+ DLBCL cells in vitro as well as in a subcutaneous DLBCL mouse model. Moreover, it shows high cytotoxic effect in CXCR4+ DLBCL cells, including induction of G2/M mitotic arrest, DNA damage, mitotic catastrophe and apoptosis. Furthermore, the nanoconjugate shows a potent reduction in lymphoma mouse dissemination without histopathological alterations in non-DLBCL infiltrated organs. Importantly, T22-AUR also exhibits lack of toxicity in human PBMCs. CONCLUSION T22-AUR exerts in vitro and in vivo anticancer effect on CXCR4+ DLBCL cells without off-target toxicity. Thus, T22-AUR promises to become an effective therapy for CXCR4+ DLBCL patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Death/drug effects
- Cell Line, Tumor
- Disease Models, Animal
- Endocytosis/drug effects
- Female
- Humans
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lysosomes/drug effects
- Lysosomes/metabolism
- Mice, Inbred NOD
- Mice, SCID
- Nanoconjugates/therapeutic use
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Receptors, CXCR4/metabolism
- Signal Transduction/drug effects
- Subcutaneous Tissue/drug effects
- Subcutaneous Tissue/pathology
- Tissue Distribution/drug effects
- Mice
Collapse
Affiliation(s)
- Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Victor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Ugutz Unzueta
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Yáiza Núñez
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
| | - Jorge Sierra
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Alberto Gallardo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Lorena Alba-Castellón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
| | - Maria Antonia Mangues
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Pharmacy, Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Patricia Álamo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Antonio Villaverde
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Esther Vázquez
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
11
|
Buschhaus JM, Humphries BA, Eckley SS, Robison TH, Cutter AC, Rajendran S, Haley HR, Bevoor AS, Luker KE, Luker GD. Targeting disseminated estrogen-receptor-positive breast cancer cells in bone marrow. Oncogene 2020; 39:5649-5662. [PMID: 32678295 PMCID: PMC7442734 DOI: 10.1038/s41388-020-01391-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/08/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022]
Abstract
Estrogen receptor-positive (ER+) breast cancer can recur up to 20 years after initial diagnosis. Delayed recurrences arise from disseminated tumors cells (DTCs) in sites such as bone marrow that remain quiescent during endocrine therapy and subsequently proliferate to produce clinically detectable metastases. Identifying therapies that eliminate DTCs and/or effectively target cells transitioning to proliferation promises to reduce risk of recurrence. To tackle this problem, we utilized a 3D co-culture model incorporating ER+ breast cancer cells and bone marrow mesenchymal stem cells to represent DTCs in a bone marrow niche. 3D co-cultures maintained cancer cells in a quiescent, viable state as measured by both single-cell and population-scale imaging. Single-cell imaging methods for metabolism by fluorescence lifetime (FLIM) of NADH and signaling by kinases Akt and ERK revealed that breast cancer cells utilized oxidative phosphorylation and signaling by Akt to a greater extent both in 3D co-cultures and a mouse model of ER+ breast cancer cells in bone marrow. Using our 3D co-culture model, we discovered that combination therapies targeting oxidative phosphorylation via the thioredoxin reductase (TrxR) inhibitor, D9, and the Akt inhibitor, MK-2206, preferentially eliminated breast cancer cells without altering viability of bone marrow stromal cells. Treatment of mice with disseminated ER+ human breast cancer showed that D9 plus MK-2206 blocked formation of new metastases more effectively than tamoxifen. These data establish an integrated experimental system to investigate DTCs in bone marrow and identify combination therapy against metabolic and kinase targets as a promising approach to effectively target these cells and reduce risk of recurrence in breast cancer.
Collapse
Affiliation(s)
- Johanna M Buschhaus
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Brock A Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Samantha S Eckley
- Unit for Laboratory Animal Medicine, University of Michigan, 412 Victor Vaughan, Ann Arbor, MI, 48109-2200, USA
- Office of Animal Resources, University of Iowa, Iowa City, IA, USA
| | - Tanner H Robison
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Alyssa C Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Shrila Rajendran
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Henry R Haley
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Avinash S Bevoor
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Gary D Luker
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
12
|
Humphries BA, Cutter AC, Buschhaus JM, Chen YC, Qyli T, Palagama DSW, Eckley S, Robison TH, Bevoor A, Chiang B, Haley HR, Sahoo S, Spinosa PC, Neale DB, Boppisetti J, Sahoo D, Ghosh P, Lahann J, Ross BD, Yoon E, Luker KE, Luker GD. Enhanced mitochondrial fission suppresses signaling and metastasis in triple-negative breast cancer. Breast Cancer Res 2020; 22:60. [PMID: 32503622 PMCID: PMC7275541 DOI: 10.1186/s13058-020-01301-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mitochondrial dynamics underlies malignant transformation, cancer progression, and response to treatment. Current research presents conflicting evidence for functions of mitochondrial fission and fusion in tumor progression. Here, we investigated how mitochondrial fission and fusion states regulate underlying processes of cancer progression and metastasis in triple-negative breast cancer (TNBC). METHODS We enforced mitochondrial fission and fusion states through chemical or genetic approaches and measured migration and invasion of TNBC cells in 2D and 3D in vitro models. We also utilized kinase translocation reporters (KTRs) to identify single cell effects of mitochondrial state on signaling cascades, PI3K/Akt/mTOR and Ras/Raf/MEK/ERK, commonly activated in TNBC. Furthermore, we determined effects of fission and fusion states on metastasis, bone destruction, and signaling in mouse models of breast cancer. RESULTS Enforcing mitochondrial fission through chemical or genetic approaches inhibited migration, invasion, and metastasis in TNBC. Breast cancer cells with predominantly fissioned mitochondria exhibited reduced activation of Akt and ERK both in vitro and in mouse models of breast cancer. Treatment with leflunomide, a potent activator of mitochondrial fusion proteins, overcame inhibitory effects of fission on migration, signaling, and metastasis. Mining existing datasets for breast cancer revealed that increased expression of genes associated with mitochondrial fission correlated with improved survival in human breast cancer. CONCLUSIONS In TNBC, mitochondrial fission inhibits cellular processes and signaling pathways associated with cancer progression and metastasis. These data suggest that therapies driving mitochondrial fission may benefit patients with breast cancer.
Collapse
Affiliation(s)
- Brock A Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Alyssa C Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Johanna M Buschhaus
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Forbes Institute for Cancer Discovery, University of Michigan, Ann Arbor, MI, USA
| | - Tonela Qyli
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Dilrukshika S W Palagama
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Samantha Eckley
- Unit for Laboratory Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tanner H Robison
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Avinash Bevoor
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Benjamin Chiang
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Henry R Haley
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Saswat Sahoo
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
| | - Phillip C Spinosa
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Dylan B Neale
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jagadish Boppisetti
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Debashis Sahoo
- Department of Pediatrics, Department of Computer Science and Engineering, Jacob's School of Engineering, Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Pradipta Ghosh
- Department of Medicine, Department of Cellular and Molecular Medicine, Rebecca and John Moore Comprehensive Cancer Center, Veterans Affairs Medical Center, University of California San Diego, La Jolla, CA, USA
| | - Joerg Lahann
- Biointerfaces Institute, Departments of Chemical Engineering, Materials Science and Engineering, Biomedical Engineering, and Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian D Ross
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Eusik Yoon
- Department of Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Deynoux M, Sunter N, Ducrocq E, Dakik H, Guibon R, Burlaud-Gaillard J, Brisson L, Rouleux-Bonnin F, le Nail LR, Hérault O, Domenech J, Roingeard P, Fromont G, Mazurier F. A comparative study of the capacity of mesenchymal stromal cell lines to form spheroids. PLoS One 2020; 15:e0225485. [PMID: 32484831 PMCID: PMC7266346 DOI: 10.1371/journal.pone.0225485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC)-spheroid models favor maintenance of stemness, ex vivo expansion and transplantation efficacy. Spheroids may also be considered as useful surrogate models of the hematopoietic niche. However, accessibility to primary cells, from bone marrow (BM) or adipose tissues, may limit their experimental use and the lack of consistency in methods to form spheroids may affect data interpretation. In this study, we aimed to create a simple model by examining the ability of cell lines, from human (HS-27a and HS-5) and murine (MS-5) BM origins, to form spheroids, compared to primary human MSCs (hMSCs). Our protocol efficiently allowed the spheroid formation from all cell types within 24 hours. Whilst hMSC-spheroids began to shrink after 24 hours, the size of spheroids from cell lines remained constant during three weeks. The difference was partially explained by the balance between proliferation and cell death, which could be triggered by hypoxia and induced oxidative stress. Our results demonstrate that, like hMSCs, MSC cell lines make reproductible spheroids that are easily handled. Thus, this model could help in understanding mechanisms involved in MSC functions and may provide a simple model by which to study cell interactions in the BM niche.
Collapse
Affiliation(s)
- Margaux Deynoux
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Nicola Sunter
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Elfi Ducrocq
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Hassan Dakik
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Roseline Guibon
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Julien Burlaud-Gaillard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Lucie Brisson
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | | | | | - Olivier Hérault
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Jorge Domenech
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Philippe Roingeard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Gaëlle Fromont
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Frédéric Mazurier
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- * E-mail:
| |
Collapse
|
14
|
Calpe B, Kovacs WJ. High-throughput screening in multicellular spheroids for target discovery in the tumor microenvironment. Expert Opin Drug Discov 2020; 15:955-967. [PMID: 32364413 DOI: 10.1080/17460441.2020.1756769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Solid tumors are highly influenced by a complex tumor microenvironment (TME) that cannot be modeled with conventional two-dimensional (2D) cell culture. In addition, monolayer culture conditions tend to induce undesirable molecular and phenotypic cellular changes. The discrepancy between in vitro and in vivo is an important factor accounting for the high failure rate in drug development. Three-dimensional (3D) multicellular tumor spheroids (MTS) more closely resemble the in vivo situation in avascularized tumors. AREAS COVERED This review describes the use of MTS for anti-cancer drug discovery, with an emphasis on high-throughput screening (HTS) compatible assays. In particular, we focus on how these assays can be used for target discovery in the context of the TME. EXPERT OPINION Arrayed MTS in microtiter plates are HTS compatible but remain more expensive and time consuming than their 2D culture counterpart. It is therefore imperative to use assays with multiplexed readouts, in order to maximize the information that can be gained with the screen. In this context, high-content screening allowing to uncover microenvironmental dependencies is the true added value of MTS-based screening compared to 2D culture-based screening. Hit translation in animal models will, however, be key to allow a broader use of MTS-based screening in industry.
Collapse
Affiliation(s)
- Blaise Calpe
- Institute of Molecular Health Sciences, ETH Zurich , Zurich, Switzerland.,Department of Biology, Debiopharm , Lausanne, Switzerland
| | - Werner J Kovacs
- Institute of Molecular Health Sciences, ETH Zurich , Zurich, Switzerland
| |
Collapse
|
15
|
Montagner M, Sahai E. In vitro Models of Breast Cancer Metastatic Dormancy. Front Cell Dev Biol 2020; 8:37. [PMID: 32195244 PMCID: PMC7062644 DOI: 10.3389/fcell.2020.00037] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Delayed relapses at distant sites are a common clinical observation for certain types of cancers after removal of primary tumor, such as breast and prostate cancer. This evidence has been explained by postulating a long period during which disseminated cancer cells (DCCs) survive in a foreign environment without developing into overt metastasis. Because of the asymptomatic nature of this phenomenon, isolation, and analysis of disseminated dormant cancer cells from clinically disease-free patients is ethically and technically highly problematic and currently these data are largely limited to the bone marrow. That said, detecting, profiling and treating indolent metastatic lesions before the onset of relapse is the imperative. To overcome this major limitation many laboratories developed in vitro models of the metastatic niche for different organs and different types of cancers. In this review we focus specifically on in vitro models designed to study metastatic dormancy of breast cancer cells (BCCs). We provide an overview of the BCCs employed in the different organotypic systems and address the components of the metastatic microenvironment that have been shown to impact on the dormant phenotype: tissue architecture, stromal cells, biochemical environment, oxygen levels, cell density. A brief description of the organ-specific in vitro models for bone, liver, and lung is provided. Finally, we discuss the strategies employed so far for the validation of the different systems.
Collapse
Affiliation(s)
- Marco Montagner
- Department of Molecular Medicine, School of Medicine and Surgery, University of Padua, Padua, Italy
| | - Erik Sahai
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
16
|
Pradhan S, Slater JH. Tunable hydrogels for controlling phenotypic cancer cell states to model breast cancer dormancy and reactivation. Biomaterials 2019; 215:119177. [PMID: 31176804 PMCID: PMC6592634 DOI: 10.1016/j.biomaterials.2019.04.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
During metastasis, disseminated tumor cells (DTCs) from the primary tumor infiltrate secondary organs and reside there for varying lengths of time prior to forming new tumors. The time delay between infiltration and active proliferation, known as dormancy, mediates the length of the latency period. DTCs may undergo one of four fates post-infiltration: death, cellular dormancy, dormant micrometastasis, or invasive growth which, is in part, mediated by extracellular matrix (ECM) properties. Recapitulation of these cell states using engineered hydrogels could facilitate the systematic and controlled investigation of the mechanisms by which ECM properties influence DTC fate. Toward this goal, we implemented a set of sixteen hydrogels with systematic variations in chemical (ligand (RGDS) density and enzymatic degradability) and mechanical (elasticity, swelling, mesh size) properties to investigate their influence on the fate of encapsulated metastatic breast cancer cells, MDA-MB-231. Cell viability, apoptosis, proliferation, metabolic activity, and morphological measurements were acquired at five-day intervals over fifteen days in culture. Analysis of the phenotypic metrics indicated the presence of four different cell states that were classified as: (1) high growth, (2) moderate growth, (3) single cell, restricted survival, dormancy, or (4) balanced dormancy. Correlating hydrogel properties with the resultant cancer cell state indicated that ligand (RGDS) density and enzymatic degradability likely had the most influence on cell fate. Furthermore, we demonstrate the ability to reactivate cells from the single cell, dormant state to the high growth state through a dynamic increase in ligand (RGDS) density after forty days in culture. This tunable engineered hydrogel platform offers insight into matrix properties regulating tumor dormancy, and the dormancy-proliferation switch, and may provide future translational benefits toward development of anti-dormancy therapeutic strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA.
| |
Collapse
|
17
|
Humphries BA, Buschhaus JM, Chen YC, Haley HR, Qyli T, Chiang B, Shen N, Rajendran S, Cutter A, Cheng YH, Chen YT, Cong J, Spinosa PC, Yoon E, Luker KE, Luker GD. Plasminogen Activator Inhibitor 1 (PAI1) Promotes Actin Cytoskeleton Reorganization and Glycolytic Metabolism in Triple-Negative Breast Cancer. Mol Cancer Res 2019; 17:1142-1154. [PMID: 30718260 DOI: 10.1158/1541-7786.mcr-18-0836] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/22/2018] [Accepted: 01/29/2019] [Indexed: 11/16/2022]
Abstract
Migration and invasion of cancer cells constitute fundamental processes in tumor progression and metastasis. Migratory cancer cells commonly upregulate expression of plasminogen activator inhibitor 1 (PAI1), and PAI1 correlates with poor prognosis in breast cancer. However, mechanisms by which PAI1 promotes migration of cancer cells remain incompletely defined. Here we show that increased PAI1 drives rearrangement of the actin cytoskeleton, mitochondrial fragmentation, and glycolytic metabolism in triple-negative breast cancer (TNBC) cells. In two-dimensional environments, both stable expression of PAI1 and treatment with recombinant PAI1 increased migration, which could be blocked with the specific inhibitor tiplaxtinin. PAI1 also promoted invasion into the extracellular matrix from coculture spheroids with human mammary fibroblasts in fibrin gels. Elevated cellular PAI1 enhanced cytoskeletal features associated with migration, actin-rich migratory structures, and reduced actin stress fibers. In orthotopic tumor xenografts, we discovered that TNBC cells with elevated PAI1 show collagen fibers aligned perpendicular to the tumor margin, an established marker of invasive breast tumors. Further studies revealed that PAI1 activates ERK signaling, a central regulator of motility, and promotes mitochondrial fragmentation. Consistent with known effects of mitochondrial fragmentation on metabolism, fluorescence lifetime imaging microscopy of endogenous NADH showed that PAI1 promotes glycolysis in cell-based assays, orthotopic tumor xenografts, and lung metastases. Together, these data demonstrate for the first time that PAI1 regulates cancer cell metabolism and suggest targeting metabolism to block motility and tumor progression. IMPLICATIONS: We identified a novel mechanism through which cancer cells alter their metabolism to promote tumor progression.
Collapse
Affiliation(s)
- Brock A Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Johanna M Buschhaus
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan.,Forbes Institute for Cancer Discovery, University of Michigan, Ann Arbor, Michigan
| | - Henry R Haley
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Tonela Qyli
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin Chiang
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Nathan Shen
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Shrila Rajendran
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Alyssa Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Yu-Heng Cheng
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan
| | - Yu-Ting Chen
- Computer Science Department UCLA, Boelter Hall, Los Angeles, California
| | - Jason Cong
- Computer Science Department UCLA, Boelter Hall, Los Angeles, California
| | - Phillip C Spinosa
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Euisik Yoon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
18
|
Liu C, Chiang B, Lewin Mejia D, Luker KE, Luker GD, Lee A. Mammary fibroblasts remodel fibrillar collagen microstructure in a biomimetic nanocomposite hydrogel. Acta Biomater 2019; 83:221-232. [PMID: 30414485 DOI: 10.1016/j.actbio.2018.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 01/22/2023]
Abstract
Architecture and microstructure of type I collagen fibers constitute central regulators of tumor invasion with aligned fibers providing a route for migration of stromal and cancer cells. Several different aspects of fibrillar collagen, such as stiffness, density, thickness, and pore size, may regulate migration of cancer cells, but determining effects of any one parameter requires clear decoupling of physical properties of collagen networks. The objective of this work is to develop and apply an in vitro three-dimensional (3D) tumor-extra cellular matrix (ECM) model with tunable physical parameters to define how stromal fibroblasts modulate collagen microstructure to control migration of breast cancer cells. We incorporated two different types of polyhedral oligomeric silsesquioxane (POSS) nano-molecules into a collagen/alginate matrix to induce different mechanisms of gelling. The resultant biomimetic, nanocomposite hydrogels show different collagen fibrillar microstructures while maintaining constant overall matrix stiffness, density, and porosimetry. Spheroids of human mammary fibroblasts embedded in these 3D matrices remodel the collagen network to varying extents based on differences in underlying matrix microstructures. The remodeled collagen matrix shows oriented, thicker fibrillar tracks, facilitating invasion of tumor cells. By decoupling effects of matrix stiffness and architecture, our nanocomposite hydrogels serve as robust platforms to investigate how biophysical properties of tumor environments control key processes regulating tumor progression in breast cancer and other malignancies. STATEMENT OF SIGNIFICANCE: Our manuscript demonstrates a new type of nanocomposite hydrogel with two different gelling mechanisms, produced by incorporating two types of polyhedral oligomeric silsesquioxane (POSS) nano-molecules into a collagen/alginate matrix. The resultant biomimetic hydrogels show different fibrillar collagen microstructures while maintaining constant overall matrix stiffness, density, and porosimetry. These gels allow us to uncouple effects of matrix stiffness versus architecture on migration and invasion of breast cancer cells and stromal fibroblasts. Upon embedding spheroids of human mammary fibroblasts (HMFs) and dissociated 231 breast cancer cells, we showed that HMFs remodeled the collagen network to differing extents dependent on starting matrix microstructures in each hydrogel. The remodeled collagen matrix showed aligned collagen fibers perpendicular to the surface of a spheroid with migrating HMFs following these fibers as occurs in tumors in vivo. To our knowledge, this is the first study showing significant different fibrillar collagen microstructures with constant collagen density and gel stiffness. This study establishes a new type of nanocomposite 3D hydrogels for studies of biophysical and cellular interactions in engineered tumor environments.
Collapse
Affiliation(s)
- Chun Liu
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Benjamin Chiang
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Daniela Lewin Mejia
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States; Department of Biomedical Engineering, University of Michigan Medical School, United States; Department of Microbiology and Immunology, University of Michigan Medical School, United States.
| | - Andre Lee
- Department of Chemical Engineering and Materials Science, Michigan State University, United States.
| |
Collapse
|
19
|
Pradhan S, Sperduto JL, Farino CJ, Slater JH. Engineered In Vitro Models of Tumor Dormancy and Reactivation. J Biol Eng 2018; 12:37. [PMID: 30603045 PMCID: PMC6307145 DOI: 10.1186/s13036-018-0120-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/16/2018] [Indexed: 12/23/2022] Open
Abstract
Metastatic recurrence is a major hurdle to overcome for successful control of cancer-associated death. Residual tumor cells in the primary site, or disseminated tumor cells in secondary sites, can lie in a dormant state for long time periods, years to decades, before being reactivated into a proliferative growth state. The microenvironmental signals and biological mechanisms that mediate the fate of disseminated cancer cells with respect to cell death, single cell dormancy, tumor mass dormancy and metastatic growth, as well as the factors that induce reactivation, are discussed in this review. Emphasis is placed on engineered, in vitro, biomaterial-based approaches to model tumor dormancy and subsequent reactivation, with a focus on the roles of extracellular matrix, secondary cell types, biochemical signaling and drug treatment. A brief perspective of molecular targets and treatment approaches for dormant tumors is also presented. Advances in tissue-engineered platforms to induce, model, and monitor tumor dormancy and reactivation may provide much needed insight into the regulation of these processes and serve as drug discovery and testing platforms.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE 19716 USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE 19716 USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE 19716 USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE 19716 USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711 USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716 USA
| |
Collapse
|
20
|
Hybrid collagen alginate hydrogel as a platform for 3D tumor spheroid invasion. Acta Biomater 2018; 75:213-225. [PMID: 29879553 DOI: 10.1016/j.actbio.2018.06.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/17/2018] [Accepted: 06/03/2018] [Indexed: 01/17/2023]
Abstract
Extracellular matrix regulates hallmark features of cancer through biochemical and mechanical signals, although mechanistic understanding of these processes remains limited by lack of models that recreate physiology of tumors. To tissue-engineer models that recapitulate three-dimensional architecture and signaling in tumors, there is a pressing need for new materials permitting flexible control of mechanical and biophysical features. We developed a hybrid hydrogel system composed of collagen and alginate to model tumor environments in breast cancer and other malignancies. Material properties of the hydrogel, including stiffness, microstructure and porosimetry, encompass parameters present in normal organs and tumors. The hydrogel possesses a well-organized, homogenous microstructure with adjustable mechanical stiffness and excellent permeability. Upon embedding multicellular tumor spheroids, we constructed a 3D tumor invasion model showing follow-the-leader migration with fibroblasts leading invasion of cancer cells similar to in vivo. We also demonstrated effects of CXCL12-CXCR4 signaling, a pathway implicated in tumor progression and metastasis, in a dual-tumor spheroid invasion model in 3D hydrogels. These studies establish a new hydrogel platform with material properties that can be tuned to investigate effects of environmental conditions on tumor progression, which will advance future studies of cancer cell invasion and response to therapy. STATEMENT OF SIGNIFICANCE Our manuscript describes a novel design of hybrid hydrogel system composed of collagen and alginate modeling 3D tumor environments in breast cancer. The hydrogel possesses a well-organized, homogenous microstructure with adjustable mechanical stiffness. Upon embedding tumor spheroids, we successfully showed a 3D tumor invasion model showing follow-the-leader migration with fibroblasts leading invasion of cancer cells similar to in vivo. To the best of our knowledge, this is the first study showing two spheroids invade simultaneously and forming bridge-like connection and effects of chemical gradients in 3D hydrogel environment. This research provides a new model for tumor-stromal interactions in cancer cell migration and establishes a novel hydrogel system for analyzing physical and biochemical signals regulating cancer progression and response to therapy.
Collapse
|
21
|
Jordahl JH, Solorio L, Sun H, Ramcharan S, Teeple CB, Haley HR, Lee KJ, Eyster TW, Luker GD, Krebsbach PH, Lahann J. 3D Jet Writing: Functional Microtissues Based on Tessellated Scaffold Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1707196. [PMID: 29484715 PMCID: PMC6112611 DOI: 10.1002/adma.201707196] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/07/2018] [Indexed: 05/21/2023]
Abstract
The advent of adaptive manufacturing techniques supports the vision of cell-instructive materials that mimic biological tissues. 3D jet writing, a modified electrospinning process reported herein, yields 3D structures with unprecedented precision and resolution offering customizable pore geometries and scalability to over tens of centimeters. These scaffolds support the 3D expansion and differentiation of human mesenchymal stem cells in vitro. Implantation of these constructs leads to the healing of critical bone defects in vivo without exogenous growth factors. When applied as a metastatic target site in mice, circulating cancer cells home in to the osteogenic environment simulated on 3D jet writing scaffolds, despite implantation in an anatomically abnormal site. Through 3D jet writing, the formation of tessellated microtissues is demonstrated, which serve as a versatile 3D cell culture platform in a range of biomedical applications including regenerative medicine, cancer biology, and stem cell biotechnology.
Collapse
Affiliation(s)
- Jacob H. Jordahl
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Luis Solorio
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Hongli Sun
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Stacy Ramcharan
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Clark B. Teeple
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Henry R. Haley
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Kyung Jin Lee
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Thomas W. Eyster
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Gary D. Luker
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Paul H. Krebsbach
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| | - Joerg Lahann
- Biointerfaces Institute, NCRC B10-A175, 2800 Plymouth Rd, Ann Arbor, MI 48109,
| |
Collapse
|
22
|
Chen YC, Humphries B, Brien R, Gibbons AE, Chen YT, Qyli T, Haley HR, Pirone ME, Chiang B, Xiao A, Cheng YH, Luan Y, Zhang Z, Cong J, Luker KE, Luker GD, Yoon E. Functional Isolation of Tumor-Initiating Cells using Microfluidic-Based Migration Identifies Phosphatidylserine Decarboxylase as a Key Regulator. Sci Rep 2018; 8:244. [PMID: 29321615 PMCID: PMC5762897 DOI: 10.1038/s41598-017-18610-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
Isolation of tumor-initiating cells currently relies on markers that do not reflect essential biologic functions of these cells. We proposed to overcome this limitation by isolating tumor-initiating cells based on enhanced migration, a function tightly linked to tumor-initiating potential through epithelial-to-mesenchymal transition (EMT). We developed a high-throughput microfluidic migration platform with automated cell tracking software and facile recovery of cells for downstream functional and genetic analyses. Using this device, we isolated a small subpopulation of migratory cells with significantly greater tumor formation and metastasis in mouse models. Whole transcriptome sequencing of migratory versus non-migratory cells from two metastatic breast cancer cell lines revealed a unique set of genes as key regulators of tumor-initiating cells. We focused on phosphatidylserine decarboxylase (PISD), a gene downregulated by 8-fold in migratory cells. Breast cancer cells overexpressing PISD exhibited reduced tumor-initiating potential in a high-throughput microfluidic mammosphere device and mouse xenograft model. PISD regulated multiple aspects of mitochondria, highlighting mitochondrial functions as therapeutic targets against cancer stem cells. This research establishes not only a novel microfluidic technology for functional isolation of tumor-initiating cells regardless of cancer type, but also a new approach to identify essential regulators of these cells as targets for drug development.
Collapse
Affiliation(s)
- Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA. .,Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA. .,Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI, 48109, USA.
| | - Brock Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Riley Brien
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Anne E Gibbons
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Yu-Ting Chen
- Computer Science Department UCLA, Boelter Hall, Los Angeles, CA, 90095-1596, USA
| | - Tonela Qyli
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Henry R Haley
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Matthew E Pirone
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Benjamin Chiang
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Annie Xiao
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Yu-Heng Cheng
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Yi Luan
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Zhixiong Zhang
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA
| | - Jason Cong
- Computer Science Department UCLA, Boelter Hall, Los Angeles, CA, 90095-1596, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA. .,Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA. .,Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI, 48109-2122, USA. .,Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
| |
Collapse
|
23
|
A Facile, In Vitro 384-Well Plate System to Model Disseminated Tumor Cells in the Bone Marrow Microenvironment. Methods Mol Biol 2018; 1686:201-213. [PMID: 29030823 DOI: 10.1007/978-1-4939-7371-2_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Bone marrow disseminated tumor cells (DTCs) are dormant cancer cells that harbor themselves in a bone marrow niche for years after patient remission before potentially returning to a proliferative state, causing recurrent cancer. DTCs reside in bone marrow environments with physiologically important mesenchymal stem cells that are often negatively affected by chemotherapy treatments. Currently, there are very few models of DTCs that recapitulate their dormant phenotype while producing enough samples to accurately quantify cancer and surrounding stromal cell behaviors. We present a three-dimensional spheroid-based model system that uses dual-color bioluminescence imaging to quantify differential cell viability in response to various compounds. We successfully screened for compounds that selectively eliminated cancer cells versus supportive stromal cells and verified results with comparison to efficacy in vivo. The spheroid coculture system successfully modeled key aspects of DTCs in the bone marrow microenvironment, facilitating testing for compounds to selectively eliminate DTCs.
Collapse
|
24
|
Abstract
Most cells in nature are not actively dividing, yet are able to return to the cell cycle given the appropriate environmental signals. There is now ample evidence that quiescent G0 cells are not shut-down but still metabolically and transcriptionally active. Quiescent cells must maintain a basal transcriptional capacity to maintain transcripts and proteins necessary for survival. This implies a tight control over RNA polymerases: RNA pol II for mRNA transcription during G0, but especially RNA pol I and RNA pol III to maintain an appropriate level of structural RNAs, raising the possibility that specific transcriptional control mechanisms evolved in quiescent cells. In accordance with this, we recently discovered that RNA interference is necessary to control RNA polymerase I transcription during G0. While this mini-review focuses on yeast model organisms (Saccharomyces cerevisiae and Schizosaccharomyces pombe), parallels are drawn to other eukaryotes and mammalian systems, in particular stem cells.
Collapse
Affiliation(s)
- Benjamin Roche
- a Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA
| | - Benoit Arcangioli
- b Genome Dynamics Unit , UMR 3525 CNRS, Institut Pasteur, 25-28 rue du Docteur Roux , Paris , France
| | - Robert Martienssen
- a Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA.,c Howard Hughes Medical Institute-Gordon and Betty Moore Foundation (HHMI-GBM) Investigator , NY , USA
| |
Collapse
|
25
|
Hinz S, Hendricks A, Wittig A, Schafmayer C, Tepel J, Kalthoff H, Becker T, Röder C. Detection of circulating tumor cells with CK20 RT-PCR is an independent negative prognostic marker in colon cancer patients - a prospective study. BMC Cancer 2017; 17:53. [PMID: 28086834 PMCID: PMC5237158 DOI: 10.1186/s12885-016-3035-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/16/2016] [Indexed: 12/01/2022] Open
Abstract
Background Detection of circulating (CTC) or disseminated tumor cells (DTC) has been associated with negative prognosis and outcome in patients with colorectal cancer, though testing for these cells is not yet part of clinical routine. There are several different methodological approaches to detect tumor cells but standardized detection assays are not implemented so far. Methods In this prospective monocentric study 299 patients with colon cancer were included. CTC and DTC were detected using CK20 RT-PCR as well as immunocytochemistry staining with anti-pan-keratin and anti-EpCAM antibodies. The primary endpoints were: Evaluation of CTC and DTC at the time of surgery and correlation with main tumor characteristics and overall (OS) and disease free survival (DFS). Results Patients with detectable CTC had a 5-year OS rate of 68% compared to a 5-year OS rate of 85% in patients without detectable CTC in the blood (p = 0.002). Detection of DTC in the bone marrow with CK20 RT-PCR was not associated with a worse OS or DFS. Detection of pan-cytokeratin positive DTC in the bone marrow correlated with a significantly reduced 5-year OS rate (p = 0.048), but detection of DTC in the bone marrow with the anti-EpCAM antibody did not significantly influence the 5-year OS rate (p = 0.958). By multivariate analyses only detection of CTC with CK20 RT-PCR in the blood was revealed to be an independent predictor of worse OS (HR1.94; 95% CI 1.0–3.7; p = 0.04) and DFS (HR 1.94; 95% CI 1.1–3.7; p = 0.044). Conclusions Detection of CTC with CK20 RT-PCR is a highly specific and independent prognostic marker in colon cancer patients. Detection of DTC in the bone marrow with CK20 RT-PCR or immunohistochemistry with anti-EpCAM antibody is not associated with a negative prognostic influence. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-3035-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sebastian Hinz
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany.
| | - Alexander Hendricks
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| | - Amke Wittig
- Division Molecular Oncology, Institute for Experimental Cancer Research, Cancer Center North, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| | - Jürgen Tepel
- Klinikum Osnabrück, Am Finkenhügel 1-3, 49076, Osnabrück, Germany
| | - Holger Kalthoff
- Division Molecular Oncology, Institute for Experimental Cancer Research, Cancer Center North, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| | - Thomas Becker
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| | - Christian Röder
- Division Molecular Oncology, Institute for Experimental Cancer Research, Cancer Center North, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller Str. 7, 24105, Kiel, Germany
| |
Collapse
|
26
|
Huang J, Woods P, Normolle D, Goff JP, Benos PV, Stehle CJ, Steinman RA. Downregulation of estrogen receptor and modulation of growth of breast cancer cell lines mediated by paracrine stromal cell signals. Breast Cancer Res Treat 2016; 161:229-243. [PMID: 27853906 DOI: 10.1007/s10549-016-4052-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
Abstract
PURPOSE Breast cancers have a poorer prognosis if estrogen receptor expression was lost during recurrence. It is unclear whether this conversion is cell autonomous or whether it can be promoted by the microenvironment during cancer dormancy. We explored the ability of marrow-derived stromal cell lines to arrest co-cultured breast cancer cells and suppress estrogen receptor alpha (ER) expression during arrest, facilitating the emergence of estrogen-independent breast cancer clones. METHODS Cancer cell growth, ER protein, microRNA, and mRNA levels were measured in breast cancer cell lines exposed to conditioned medium from marrow stromal lines in the presence and absence of estrogen and of signaling pathway modulators. RESULTS We demonstrate that paracrine signaling from the stromal cell line HS5 downregulated ER in T47D and MCF7 breast cancer cells. This occurred at the mRNA level and also through decreased ER protein stability. Additionally, conditioned medium (CM) from HS5 arrested the breast cancer cells in G0/G1 in part through interleukin-1 (IL1) and inhibited cancer cell growth despite the activation of proliferative pathways (Erk and AKT) by the CM. Similar findings were observed for CM from the hFOB 1.19 osteoblastic cell line but not from two other fibroblastic marrow lines, HS27A and KM101. HS5-CM inhibition of MCF7 proliferation could not be restored by exogenous ER, but was restored by the IL1-antagonist IL1RA. In the presence of IL1RA, HS5-CM activation of AKT and Erk enabled the outgrowth of breast cancer cells with suppressed ER that were fulvestrant-resistant and estrogen-independent. CONCLUSIONS We conclude that marrow-derived stromal cells can destabilize estrogen receptor protein to convert the ER status of growth-arrested ER+ breast cancer cell lines. The balance between stromal pro- and anti-proliferative signals controlled the switch from a dormant phenotype to estrogen-independent cancer cell growth.
Collapse
Affiliation(s)
- J Huang
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Suite 2.26f Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.,School of Medicine, Tsinghua University, Beijing, China
| | - P Woods
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Suite 2.26f Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - D Normolle
- Department of Biostatistics, University of Pittsburgh and University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - J P Goff
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Suite 2.26f Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - P V Benos
- Department of Computational Biology, University of Pittsburgh and University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - C J Stehle
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Suite 2.26f Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - R A Steinman
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Suite 2.26f Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA. .,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
27
|
In vivo targeting of metastatic breast cancer via tumor vasculature-specific nano-graphene oxide. Biomaterials 2016; 104:361-71. [PMID: 27490486 DOI: 10.1016/j.biomaterials.2016.07.029] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/20/2016] [Accepted: 07/23/2016] [Indexed: 12/21/2022]
Abstract
Angiogenesis, i.e. the formation of neovasculatures, is a critical process during cancer initiation, progression, and metastasis. Targeting of angiogenic markers on the tumor vasculature can result in more efficient delivery of nanomaterials into tumor since no extravasation is required. Herein we demonstrated efficient targeting of breast cancer metastasis in an experimental murine model with nano-graphene oxide (GO), which was conjugated to a monoclonal antibody (mAb) against follicle-stimulating hormone receptor (FSHR). FSHR has been confirmed to be a highly selective tumor vasculature marker, which is abundant in both primary and metastatic tumors. These functionalized GO nano-conjugates had diameters of ∼120 nm based on atomic force microscopy (AFM), TEM, and dynamic laser scattering (DLS) measurement. (64)Cu was incorporated as a radiolabel which enabled the visualization of these GO conjugates by positron emission tomography (PET) imaging. Breast cancer lung metastasis model was established by intravenous injection of click beetle green luciferase-transfected MDA-MB-231 (denoted as cbgLuc-MDA-MB-231) breast cancer cells into female nude mice and the tumor growth was monitored by bioluminescence imaging (BLI). Systematic in vitro and in vivo studies have been performed to investigate the stability, targeting efficacy and specificity, and tissue distribution of GO conjugates. Flow cytometry and fluorescence microscopy examination confirmed the targeting specificity of FSHR-mAb attached GO conjugates against cellular FSHR. More potent and persistent uptake of (64)Cu-NOTA-GO-FSHR-mAb in cbgLuc-MDA-MB-231 nodules inside the lung was witnessed when compared with that of non-targeted GO conjugates ((64)Cu-NOTA-GO). Histology evaluation also confirmed the vasculature accumulation of GO-FSHR-mAb conjugates in tumor at early time points while they were non-specifically captured in liver and spleen. In addition, these GO conjugates can serve as good drug carriers with satisfactory drug loading capacity (e.g. for doxorubicin [DOX], 756 mg/g). Enhanced drug delivery efficiency in cbgLuc-MDA-MB-231 metastatic sites was demonstrated in DOX-loaded GO-FSHR-mAb by fluorescence imaging. This FSHR-targeted, GO-based nanoplatform can serve as a useful tool for early metastasis detection and targeted delivery of therapeutics.
Collapse
|
28
|
Cavnar SP, Xiao A, Gibbons AE, Rickelmann AD, Neely T, Luker KE, Takayama S, Luker GD. Imaging Sensitivity of Quiescent Cancer Cells to Metabolic Perturbations in Bone Marrow Spheroids. Tomography 2016; 2:146-157. [PMID: 27478871 PMCID: PMC4963031 DOI: 10.18383/j.tom.2016.00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Malignant cells from breast cancer and other common cancers such as prostate and melanoma may persist in bone marrow as quiescent, non-dividing cells that remain viable for years or even decades before resuming proliferation to cause recurrent disease. This phenomenon, referred to clinically as tumor dormancy, poses tremendous challenges to curing patients with breast cancer. Quiescent tumor cells resist chemotherapy drugs that predominantly target proliferating cells, limiting success of neo-adjuvant and adjuvant therapies. We recently developed a 3D spheroid model of quiescent breast cancer cells in bone marrow for mechanistic and drug testing studies. We combined this model with optical imaging methods for label-free detection of cells preferentially utilizing glycolysis versus oxidative metabolism to investigate the metabolic state of co-culture spheroids with different bone marrow stromal and breast cancer cells. Through imaging and biochemical assays, we identified different metabolic states of bone marrow stromal cells that control metabolic status and flexibilities of co-cultured breast cancer cells. We tested metabolic stresses and targeted inhibition of specific metabolic pathways to identify approaches to preferentially eliminate quiescent breast cancer cells from bone marrow environments. These studies establish an integrated imaging approach to analyze metabolism in complex tissue environments to identify new metabolically-targeted cancer therapies.
Collapse
Affiliation(s)
- Stephen P. Cavnar
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
| | - Annie Xiao
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Anne E. Gibbons
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew D. Rickelmann
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Taylor Neely
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kathryn E. Luker
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Biointerfaces Institute, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
| | - Gary D. Luker
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan
- Department of Radiology, Center for Molecular Imaging, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Macromolecular Science and Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan; and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
29
|
Ham SL, Joshi R, Thakuri PS, Tavana H. Liquid-based three-dimensional tumor models for cancer research and drug discovery. Exp Biol Med (Maywood) 2016; 241:939-54. [PMID: 27072562 PMCID: PMC4950350 DOI: 10.1177/1535370216643772] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumors are three-dimensional tissues where close contacts between cancer cells, intercellular interactions between cancer and stromal cells, adhesion of cancer cells to the extracellular matrix, and signaling of soluble factors modulate functions of cancer cells and their response to therapeutics. Three-dimensional cultures of cancer cells overcome limitations of traditionally used monolayer cultures and recreate essential characteristics of tumors such as spatial gradients of oxygen, growth factors, and metabolites and presence of necrotic, hypoxic, quiescent, and proliferative cells. As such, three-dimensional tumor models provide a valuable tool for cancer research and oncology drug discovery. Here, we describe different tumor models and primarily focus on a model known as tumor spheroid. We summarize different technologies of spheroid formation, and discuss the use of spheroids to address the influence of stromal fibroblasts and immune cells on cancer cells in tumor microenvironment, study cancer stem cells, and facilitate compound screening in the drug discovery process. We review major techniques for quantification of cellular responses to drugs and discuss challenges ahead to enable broad utility of tumor spheroids in research laboratories, integrate spheroid models into drug development and discovery pipeline, and use primary tumor cells for drug screening studies to realize personalized cancer treatment.
Collapse
Affiliation(s)
- Stephanie L Ham
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325, USA
| | - Pradip S 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
|