1
|
Fuochi S, Galligioni V. Disease Animal Models for Cancer Research. Methods Mol Biol 2023; 2645:105-125. [PMID: 37202613 DOI: 10.1007/978-1-0716-3056-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite nonanimal methods (NAMs) are more and more exploited and new NAMs are developed and validated, animal models are still used in cancer research. Animals are used at multiple levels, from understanding molecular traits and pathways, to mimicking clinical aspects of tumor progression, to drug testing. In vivo approaches are not trivial and involve cross-disciplinary knowledge: animal biology and physiology, genetics, pathology, and animal welfare.The aim of this chapter is not to list and address all animal models used in cancer research. Instead, the authors would like to guide experimenters in the strategies to adopt in both planning and performing in vivo experimental procedures, including the choice of cancer animal models.
Collapse
Affiliation(s)
- Sara Fuochi
- Universität Bern, Experimental Animal Center, Bern, Switzerland
| | - Viola Galligioni
- Netherlands Institute for Neuroscience - KNAW, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Rojas JD, Joiner JB, Velasco B, Bautista KJB, Aji AM, Moore CJ, Beaumont NJ, Pylayeva-Gupta Y, Dayton PA, Gessner RC, Czernuszewicz TJ. Validation of a combined ultrasound and bioluminescence imaging system with magnetic resonance imaging in orthotopic pancreatic murine tumors. Sci Rep 2022; 12:102. [PMID: 34996939 PMCID: PMC8741958 DOI: 10.1038/s41598-021-03684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/08/2021] [Indexed: 11/30/2022] Open
Abstract
Preclinical mouse solid tumor models are widely used to evaluate efficacy of novel cancer therapeutics. Recent reports have highlighted the need for utilizing orthotopic implantation to represent clinical disease more accurately, however the deep tissue location of these tumors makes longitudinal assessment challenging without the use of imaging techniques. The purpose of this study was to evaluate the performance of a new multi-modality high-throughput in vivo imaging system that combines bioluminescence imaging (BLI) with robotic, hands-free ultrasound (US) for evaluating orthotopic mouse models. Long utilized in cancer research as independent modalities, we hypothesized that the combination of BLI and US would offer complementary advantages of detection sensitivity and quantification accuracy, while mitigating individual technological weaknesses. Bioluminescent pancreatic tumor cells were injected into the pancreas tail of C57BL/6 mice and imaged weekly with the combination system and magnetic resonance imaging (MRI) to serve as a gold standard. BLI photon flux was quantified to assess tumor activity and distribution, and US and MRI datasets were manually segmented for gross tumor volume. Robotic US and MRI demonstrated a strong agreement (R2 = 0.94) for tumor volume measurement. BLI showed a weak overall agreement with MRI (R2 = 0.21), however, it offered the greatest sensitivity to detecting the presence of tumors. We conclude that combining BLI with robotic US offers an efficient screening tool for orthotopic tumor models.
Collapse
Affiliation(s)
| | - Jordan B Joiner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Brian Velasco
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Kathlyne Jayne B Bautista
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | | | | | | | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | - Tomasz J Czernuszewicz
- SonoVol, Inc, Durham, NC, USA.
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Thaiss WM, Gatidis S, Sartorius T, Machann J, Peter A, Eigentler TK, Nikolaou K, Pichler BJ, Kneilling M. Noninvasive, longitudinal imaging-based analysis of body adipose tissue and water composition in a melanoma mouse model and in immune checkpoint inhibitor-treated metastatic melanoma patients. Cancer Immunol Immunother 2020; 70:1263-1275. [PMID: 33130917 PMCID: PMC8053172 DOI: 10.1007/s00262-020-02765-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022]
Abstract
Background As cancer cachexia (CC) is associated with cancer progression, early identification would be beneficial. The aim of this study was to establish a workflow for automated MRI-based segmentation of visceral (VAT) and subcutaneous adipose tissue (SCAT) and lean tissue water (LTW) in a B16 melanoma animal model, monitor diseases progression and transfer the protocol to human melanoma patients for therapy assessment. Methods For in vivo monitoring of CC B16 melanoma-bearing and healthy mice underwent longitudinal three-point DIXON MRI (days 3, 12, 17 after subcutaneous tumor inoculation). In a prospective clinical study, 18 metastatic melanoma patients underwent MRI before, 2 and 12 weeks after onset of checkpoint inhibitor therapy (CIT; n = 16). We employed an in-house MATLAB script for automated whole-body segmentation for detection of VAT, SCAT and LTW. Results B16 mice exhibited a CC phenotype and developed a reduced VAT volume compared to baseline (B16 − 249.8 µl, − 25%; controls + 85.3 µl, + 10%, p = 0.003) and to healthy controls. LTW was increased in controls compared to melanoma mice. Five melanoma patients responded to CIT, 7 progressed, and 6 displayed a mixed response. Responding patients exhibited a very limited variability in VAT and SCAT in contrast to others. Interestingly, the LTW was decreased in CIT responding patients (− 3.02% ± 2.67%; p = 0.0034) but increased in patients with progressive disease (+ 1.97% ± 2.19%) and mixed response (+ 4.59% ± 3.71%). Conclusion MRI-based segmentation of fat and water contents adds essential additional information for monitoring the development of CC in mice and metastatic melanoma patients during CIT or other treatment approaches.
Collapse
Affiliation(s)
- Wolfgang M Thaiss
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany.,Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany.,Department of Nuclear Medicine, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany.,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany
| | - Tina Sartorius
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany.,Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peter
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany.,Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstreet 20, 72076, Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany.,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076, Tübingen, Germany
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany. .,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany. .,Department of Dermatology, University Hospital Tübingen, Liebermeisterstreet 20, 72076, Tübingen, Germany.
| |
Collapse
|
4
|
Taschauer A, Polzer W, Alioglu F, Billerhart M, Decker S, Kittelmann T, Geppl E, Elmenofi S, Zehl M, Urban E, Sami H, Ogris M. Peptide-Targeted Polyplexes for Aerosol-Mediated Gene Delivery to CD49f-Overexpressing Tumor Lesions in Lung. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:774-786. [PMID: 31734558 PMCID: PMC6861568 DOI: 10.1016/j.omtn.2019.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 10/03/2019] [Accepted: 10/12/2019] [Indexed: 12/16/2022]
Abstract
Peptide ligands can enhance delivery of nucleic acid-loaded nanoparticles to tumors by promoting their cell binding and internalization. Lung tumor lesions accessible from the alveolar side can be transfected, in principle, using gene vectors delivered as an aerosol. The cell surface marker CD49f (Integrin α6) is frequently upregulated in metastasizing, highly aggressive tumors. In this study, we utilize a CD49f binding peptide coupled to linear polyethylenimine (LPEI) promoting gene delivery into CD49f-overexpressing tumor cells in vitro and into lung lesions in vivo. We have synthesized a molecular conjugate based on LPEI covalently attached to the CD49f binding peptide CYESIKVAVS via a polyethylene glycol (PEG) spacer. Particles formed with plasmid DNA were small (<200 nm) and could be aerosolized without causing major aggregation or particle loss. In vitro, CD49f targeting significantly improved plasmid uptake and reporter gene expression on both human and murine tumor cell lines. For evaluation in vivo, localization and morphology of 4T1 murine triple-negative breast cancer tumor lesions in the lung of syngeneic BALB/c mice were identified by MRI. Polyplexes applied via intratracheal aerosolization were well tolerated and resulted in measurable transgene activity of the reporter gene firefly luciferase in tumor areas by bioluminescence imaging (BLI). Transfectability of tumors correlated with their accessibility for the aerosol. With CD49f-targeted polyplexes, luciferase activity was considerably increased and was restricted to the tumor area.
Collapse
Affiliation(s)
- Alexander Taschauer
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Wolfram Polzer
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Fatih Alioglu
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Magdalena Billerhart
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Simon Decker
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Theresa Kittelmann
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Emanuela Geppl
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Salma Elmenofi
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Martin Zehl
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Währingerstrasse 38, 1090 Vienna, Austria
| | - Ernst Urban
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Haider Sami
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| | - Manfred Ogris
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| |
Collapse
|
5
|
Dieterich LC, Kapaklikaya K, Cetintas T, Proulx ST, Commerford CD, Ikenberg K, Bachmann SB, Scholl J, Detmar M. Transcriptional profiling of breast cancer-associated lymphatic vessels reveals VCAM-1 as regulator of lymphatic invasion and permeability. Int J Cancer 2019; 145:2804-2815. [PMID: 31344266 PMCID: PMC6771758 DOI: 10.1002/ijc.32594] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/11/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022]
Abstract
Tumor‐associated lymphangiogenesis and lymphatic invasion of tumor cells correlate with poor outcome in many tumor types, including breast cancer. Various explanations for this correlation have been suggested in the past, including the promotion of lymphatic metastasis and an immune‐inhibitory function of lymphatic endothelial cells (LECs). However, the molecular features of tumor‐associated lymphatic vessels and their implications for tumor progression have been poorly characterized. Here, we report the first transcriptional analysis of tumor‐associated LECs directly isolated from the primary tumor in an orthotopic mouse model of triple negative breast cancer (4T1). Gene expression analysis showed a strong upregulation of inflammation‐associated genes, including endothelial adhesion molecules such as VCAM‐1, in comparison to LECs derived from control tissue. In vitro experiments demonstrated that VCAM‐1 is not involved in the adhesion of tumor cells to LECs but unexpectedly promoted lymphatic permeability by weakening of lymphatic junctions, most likely through a mechanism triggered by interactions with integrin α4 which was also induced in tumor‐associated LECs. In line with this, in vivo blockade of VCAM‐1 reduced lymphatic invasion of 4T1 cells. Taken together, our findings suggest that disruption of lymphatic junctions and increased permeability via tumor‐induced lymphatic VCAM‐1 expression may represent a new target to block lymphatic invasion and metastasis. What's new? Tumor‐associated lymphatic vessels serve important roles in tumor progression and metastasis. Nonetheless, little is known about the molecular changes in these vessels that give rise to a tumor‐promoting phenotype. In this study, transcriptional analysis was performed on lymphatic endothelial cells (LECs) isolated from a mouse model of triple‐negative breast cancer. Endothelial adhesion molecules, including tumor‐induced VCAM‐1, were strongly upregulated in tumor‐associated LECs. Additional experiments showed that VCAM‐1 upregulation influences lymphatic permeability and that its inhibition attenuates lymphatic breast cancer cell invasion. The findings identify VCAM‐1 as a potential target for the blockade of lymphatic invasion of tumor cells.
Collapse
Affiliation(s)
- Lothar C. Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Kübra Kapaklikaya
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Timur Cetintas
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Steven T. Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Catharina D. Commerford
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Kristian Ikenberg
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Samia B. Bachmann
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Jeannette Scholl
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| |
Collapse
|
6
|
Rego GNDA, Mamani JB, Souza TKF, Nucci MP, Silva HRD, Gamarra LF. Therapeutic evaluation of magnetic hyperthermia using Fe3O4-aminosilane-coated iron oxide nanoparticles in glioblastoma animal model. EINSTEIN-SAO PAULO 2019; 17:eAO4786. [PMID: 31390427 PMCID: PMC6668731 DOI: 10.31744/einstein_journal/2019ao4786] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/28/2019] [Indexed: 11/21/2022] Open
Abstract
Objective: To evaluate the potential of magnetic hyperthermia using aminosilane-coated superparamagnetic iron oxide nanoparticles in glioblastoma tumor model. Methods: The aminosilane-coated superparamagnetic iron oxide nanoparticles were analyzed as to their stability in aqueous medium and their heating potential through specific absorption rate, when submitted to magnetic hyperthermia with different frequencies and intensities of alternating magnetic field. In magnetic hyperthermia in vitro assays, the C6 cells cultured and transduced with luciferase were analyzed by bioluminescence in the absence/presence of alternating magnetic field, and also with and without aminosilane-coated superparamagnetic iron oxide nanoparticles. In the in vivo study, the measurement of bioluminescence was performed 21 days after glioblastoma induction with C6 cells in rats. After 24 hours, the aminosilane-coated superparamagnetic iron oxide nanoparticles were implanted in animals, and magnetic hyperthermia was performed for 40 minutes, using the best conditions of frequency and intensity of alternating magnetic field tested in the in vitro study (the highest specific absorption rate value) and verified the difference of bioluminescence before and after magnetic hyperthermia. Results: The aminosilane-coated superparamagnetic iron oxide nanoparticles were stable, and their heating capacity increased along with higher frequency and intensity of alternating magnetic field. The magnetic hyperthermia application with 874kHz and 200 Gauss of alternating magnetic field determined the best value of specific absorption rate (194.917W/g). When these magnetic hyperthermia parameters were used in in vitro and in vivo analysis, resulted in cell death of 52.0% and 32.8%, respectively, detected by bioluminescence. Conclusion: The magnetic hyperthermia was promissing for the therapeutical process of glioblastoma tumors in animal model, using aminosilane-coated superparamagnetic iron oxide nanoparticles, which presented high specific absorption rate.
Collapse
Affiliation(s)
| | | | | | - Mariana Penteado Nucci
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
7
|
Ravoori MK, Margalit O, Singh S, Kim SH, Wei W, Menter DG, DuBois RN, Kundra V. Magnetic Resonance Imaging and Bioluminescence Imaging for Evaluating Tumor Burden in Orthotopic Colon Cancer. Sci Rep 2019; 9:6100. [PMID: 30988343 PMCID: PMC6465293 DOI: 10.1038/s41598-019-42230-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022] Open
Abstract
Quantifying tumor burden is important for following the natural history of orthotopic colon cancer and therapeutic efficacy. Bioluminescence imaging (BLI) is commonly used for such assessment and has both advantages and limitations. We compared BLI and magnetic resonance imaging (MRI) for quantifying orthotopic tumors in a mouse model of colon cancer. Among sequences tested, T2-based MRI imaging ranked best overall for colon cancer border delineation, contrast, and conspicuity. Longitudinal MRI detected tumor outside the colon, indistinguished by BLI. Colon tumor weights calculated from MRI in vivo correlated highly with tumor weights measured ex vivo whereas the BLI signal intensities correlated relatively poorly and this difference in correlations was highly significant. This suggests that MRI may more accurately assess tumor burden in longitudinal monitoring of orthotopic colon cancer in this model as well as in other models.
Collapse
Affiliation(s)
- M K Ravoori
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA
| | - O Margalit
- Department of Oncology, Chaim Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-HaShomer, 52621, Israel
| | - S Singh
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA
| | - Sun-Hee Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA
| | - W Wei
- Department of Biostatistics, U.T.-M.D. Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA
| | - D G Menter
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, U.T.-M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - R N DuBois
- MUSC College of Medicine, Dean's Office, 96 Jonathan Lucas Street, Suite 601, MSC 617, Charleston, SC, 29425, USA
| | - V Kundra
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA. .,Department of Radiology, U.T.-M.D. Anderson Cancer Center, 1400 Pressler St., Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Grudzinski J, Marsh I, Titz B, Jeffery J, Longino M, Kozak K, Lange K, Larrabee J, Weichmann A, Moser A, Bednarz B. CLR 125 Auger Electrons for the Targeted Radiotherapy of Triple-Negative Breast Cancer. Cancer Biother Radiopharm 2018; 33:87-95. [PMID: 29641256 DOI: 10.1089/cbr.2017.2376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Auger electrons emitted by radioisotopes such as 125I have a high linear energy transfer and short mean-free path in tissue (<10 μm), making them suitable for treating micrometastases while sparing normal tissues. The authors developed and subsequently investigated a cancer cell-selective small molecule phospholipid ether analog to deliver 125I to triple-negative breast cancer (TNBC) cells in vivo. METHODS A Current Good Manufacturing Practice (cGMP) method to radiolabel 125I-CLR1404 (CLR 125) with >95% radiochemical purity was established. To estimate CLR 125 in vivo dosimetry and identify dose-limiting organs, the biodistribution of the analog compound 124I-CLR1404 (CLR 124) was investigated using micro-positron emission tomography (PET)/computed tomography (CT) in conjunction with a Monte Carlo dosimetry platform to estimate CLR 125 dosimetry. In vivo antitumor efficacy was tested by injecting nude mice bearing either MDA-MB-231-luc orthotopic xenografts or lung metastases with 74 MBq (3.7 GBq/kg) of CLR 125 or an equivalent mass amount of nonradiolabeled CLR 125. Longitudinal tumor measurements using calipers and bioluminescence imaging were obtained for the xenografts and lung metastases, respectively. RESULTS Dosimetry analysis estimated that CLR 125 would impart the largest absorbed dose to the tumor per injected activity (0.261 ± 0.023 Gy/MBq) while the bone marrow, which is generally the dose-limiting organ for CLR1404, appears to have the lowest (0.063 ± 0.005 Gy/MBq). At administered activities of up to 74 MBq (3.7 GBq/kg), mice did not experience signs of toxicity. In addition, a single dose of CLR 125 reduced the volume of orthotopic primary TNBC xenografts by ∼60% compared to control vehicle (p < 0.001) and significantly extended survival. In addition, CLR 125 was efficacious against preclinical metastatic TNBC models by inhibiting the progression of micrometastases (p < 0.01). CONCLUSIONS Targeted radionuclide therapy with CLR 125 displayed significant antitumor efficacy in vivo, suggesting promise for treatment of TNBC micrometastases.
Collapse
Affiliation(s)
- Joseph Grudzinski
- 1 Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin
| | - Ian Marsh
- 1 Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin
| | | | - Justin Jeffery
- 3 University of Wisconsin Carbone Cancer Center , Madison, Wisconsin
| | - Marc Longino
- 2 Cellectar Biosciences, Inc. , Madison, Wisconsin
| | - Kevin Kozak
- 2 Cellectar Biosciences, Inc. , Madison, Wisconsin
| | | | | | - Ashley Weichmann
- 3 University of Wisconsin Carbone Cancer Center , Madison, Wisconsin
| | - Amy Moser
- 3 University of Wisconsin Carbone Cancer Center , Madison, Wisconsin.,4 Department of Human Oncology, University of Wisconsin-Madison , Madison, Wisconsin
| | - Bryan Bednarz
- 1 Department of Medical Physics, University of Wisconsin-Madison , Madison, Wisconsin.,3 University of Wisconsin Carbone Cancer Center , Madison, Wisconsin.,4 Department of Human Oncology, University of Wisconsin-Madison , Madison, Wisconsin
| |
Collapse
|
9
|
Fricke IB, De Souza R, Costa Ayub L, Francia G, Kerbel R, Jaffray DA, Zheng J. Spatiotemporal assessment of spontaneous metastasis formation using multimodal in vivo imaging in HER2+ and triple negative metastatic breast cancer xenograft models in mice. PLoS One 2018; 13:e0196892. [PMID: 29723251 PMCID: PMC5933713 DOI: 10.1371/journal.pone.0196892] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/20/2018] [Indexed: 12/31/2022] Open
Abstract
Background Preclinical breast cancer models recapitulating the clinical course of metastatic disease are crucial for drug development. Highly metastatic cell lines forming spontaneous metastasis following orthotopic implantation were previously developed and characterized regarding their biological and histological characteristics. This study aimed to non-invasively and longitudinally characterize the spatiotemporal pattern of metastasis formation and progression in the MDA-MB-231-derived triple negative LM2-4 and HER2+ LM2-4H2N cell lines, using bioluminescence imaging (BLI), contrast enhanced computed tomography (CT), fluorescence imaging, and 2-deoxy-2-[fluorine-18]fluoro-D-glucose positron emission tomography ([18F]FDG-PET). Material and methods LM2-4, LM2-4H2N, and MDA-MB-231 tumors were established in the right inguinal mammary fat pad (MFP) of female SCID mice and resected 14–16 days later. Metastasis formation was monitored using BLI. Metabolic activity of primary and metastatic lesions in mice bearing LM2-4 or LM2-4H2N was assessed by [18F]FDG-PET. Metastatic burden at study endpoint was assessed by CT and fluorescence imaging following intravenous dual-modality liposome agent administration. Results Comparable temporal metastasis patterns were observed using BLI for the highly metastatic cell lines LM2-4 and LM2-4H2N, while metastasis formed about 10 days later for MDA-MB-231. 21 days post primary tumor resection, metastases were detected in 86% of LM2-4, 69% of LM2-4H2N, and 60% of MDA-MB-231 inoculated mice, predominantly in the axillary region, contralateral MFP, and liver/lung. LM2-4 and LM2-4H2N tumors displayed high metabolism based on [18F]FDG-PET uptake. Lung metastases were detected as the [18F]FDG-PET uptake increased significantly between pre- and post-metastasis scan. Using a liposomal dual-modality agent, CT and fluorescence confirmed BLI detected lesions and identified additional metastatic nodules in the intraperitoneal cavity and lung. Conclusions The combination of complementary anatomical and functional imaging techniques can provide high sensitivity characterization of metastatic disease spread, progression and overall disease burden. The described models and imaging toolset can be implemented as an effective means for quantitative treatment response evaluation in metastatic breast cancer.
Collapse
Affiliation(s)
- Inga B. Fricke
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| | - Raquel De Souza
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Lais Costa Ayub
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Giulio Francia
- Biological Sciences Platform, Sunnybrook Research Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Robert Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - David A. Jaffray
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Jinzi Zheng
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Baklaushev VP, Kilpeläinen A, Petkov S, Abakumov MA, Grinenko NF, Yusubalieva GM, Latanova AA, Gubskiy IL, Zabozlaev FG, Starodubova ES, Abakumova TO, Isaguliants MG, Chekhonin VP. Luciferase Expression Allows Bioluminescence Imaging But Imposes Limitations on the Orthotopic Mouse (4T1) Model of Breast Cancer. Sci Rep 2017; 7:7715. [PMID: 28798322 PMCID: PMC5552689 DOI: 10.1038/s41598-017-07851-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/04/2017] [Indexed: 01/08/2023] Open
Abstract
Implantation of reporter-labeled tumor cells in an immunocompetent host involves a risk of their immune elimination. We have studied this effect in a mouse model of breast cancer after the orthotopic implantation of mammary gland adenocarcinoma 4T1 cells genetically labelled with luciferase (Luc). Mice were implanted with 4T1 cells and two derivative Luc-expressing clones 4T1luc2 and 4T1luc2D6 exhibiting equal in vitro growth rates. In vivo, the daughter 4T1luc2 clone exhibited nearly the same, and 4T1luc2D6, a lower growth rate than the parental cells. The metastatic potential of 4T1 variants was assessed by magnetic resonance, bioluminescent imaging, micro-computed tomography, and densitometry which detected 100-μm metastases in multiple organs and bones at the early stage of their development. After 3-4 weeks, 4T1 generated 11.4 ± 2.1, 4T1luc2D6, 4.5 ± 0.6; and 4T1luc2, <1 metastases per mouse, locations restricted to lungs and regional lymph nodes. Mice bearing Luc-expressing tumors developed IFN-γ response to the dominant CTL epitope of Luc. Induced by intradermal DNA-immunization, such response protected mice from the establishment of 4T1luc2-tumors. Our data show that natural or induced cellular response against the reporter restricts growth and metastatic activity of the reporter-labelled tumor cells. Such cells represent a powerful instrument for improving immunization technique for cancer vaccine applications.
Collapse
Affiliation(s)
- V P Baklaushev
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Biomedical Agency of the Russian Federation, Moscow, Russia.
| | - A Kilpeläinen
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - S Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M A Abakumov
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N F Grinenko
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - G M Yusubalieva
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Latanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia
| | - I L Gubskiy
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - F G Zabozlaev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Biomedical Agency of the Russian Federation, Moscow, Russia
| | - E S Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia
| | - T O Abakumova
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M G Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Chumakov Federal Scientific Center for Research and Development of Immunobiological Preparations, Moscow, Russia.
- N.F. Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia.
- Riga Stradins University, Riga, Latvia.
| | - V P Chekhonin
- Research and Education Center for Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Fundamental and Applied Neurobiology, Serbsky National Research Center for Social and Forensic Psychiatry, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
11
|
Nedrow JR, Josefsson A, Park S, Ranka S, Roy S, Sgouros G. Imaging of Programmed Cell Death Ligand 1: Impact of Protein Concentration on Distribution of Anti-PD-L1 SPECT Agents in an Immunocompetent Murine Model of Melanoma. J Nucl Med 2017; 58:1560-1566. [PMID: 28522738 DOI: 10.2967/jnumed.117.193268] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/26/2017] [Indexed: 12/25/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is part of an immune checkpoint system that is essential for preventing autoimmunity and cancer. Recent approaches in immunotherapy that target immune checkpoints have shown great promise in a variety of cancers, including metastatic melanoma. The use of targeted molecular imaging would help identify patients who will best respond to anti-PD-L1 treatment while potentially providing key information to limit immune-related adverse effects. Recently, we developed an antibody-based PD-L1-targeted SPECT agent-111In-diethylenetriaminepentaacetic acid (DTPA)-anti-PD-L1-to identify PD-L1-positive tumors in vivo. To best use such PD-L1-targeted imaging agents, it is important, as a first step, to understand how the signal is affected by different parameters. Methods: We evaluated the impact of protein concentration on the distribution of 111In-DTPA-anti-PD-L1 in a murine model of aggressive melanoma. Results:111In-DTPA-anti-PD-L1 (dissociation constant, 0.6 ± 0.1 nM) demonstrated increased uptake in B16F10 tumors at protein concentrations equaling or exceeding 1 mg/kg at 24 h and 3 mg/kg at 72 h. At 24 h, the PD-L1-rich spleen and lungs demonstrated decreasing uptake with increasing protein concentration. At 72 h, uptake in the thymus was significantly increased at protein concentrations of 3 mg/kg or greater. Both time points demonstrated increased tracer amounts remaining in circulation as the amount of cold antibody was increased. Conclusion: These studies demonstrate that 111In-DTPA-anti-PD-L1 is capable of identifying tumors that overexpresses PD-L1 and monitoring the impact of PD-L1-rich organs on the distribution of anti-PD-L1 antibodies.
Collapse
Affiliation(s)
- Jessie R Nedrow
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anders Josefsson
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sunju Park
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sagar Ranka
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sanchita Roy
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George Sgouros
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Paczuska J, Świtalska M, Nowak M, Kiełbowicz Z. Effectiveness of CO2laser in an experimental mammary gland adenocarcinoma model. Vet Comp Oncol 2017; 16:47-54. [DOI: 10.1111/vco.12310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 01/04/2017] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- J. Paczuska
- Department of Surgery, Faculty of Veterinary Medicine; Wrocław University of Environmental and Life Sciences; Wrocław Poland
| | - M. Świtalska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; Wrocław Poland
| | - M. Nowak
- Department of Pathology; Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences; Wrocław Poland
| | - Z. Kiełbowicz
- Department of Surgery, Faculty of Veterinary Medicine; Wrocław University of Environmental and Life Sciences; Wrocław Poland
| |
Collapse
|
13
|
Longitudinal microcomputed tomography-derived biomarkers for lung metastasis detection in a syngeneic mouse model: added value to bioluminescence imaging. J Transl Med 2017; 97:24-33. [PMID: 27869796 DOI: 10.1038/labinvest.2016.114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 01/22/2023] Open
Abstract
With more patients dying from metastasis than from primary cancers, metastasis is a very important area in cancer research. Investigators thereby heavily rely on animal models of metastasis to common organs such as the lung to improve our insight into the pathogenesis and to research novel therapeutic approaches to combat metastasis. In this experimental context, novel tools that allow longitudinal monitoring of lung metastasis in individual animals are highly needed. We have therefore evaluated for the first time microcomputed tomography (μCT) as a very efficient and crossvalidated means to noninvasively and repeatedly monitor metastasis to the lung in individual, free-breathing syngeneic mice. Two individual clones of KLN205 cancer cells were intravenously injected in syngeneic DBA/2 mice and lung metastasis was monitored weekly during 3 weeks using μCT, and was compared with the current gold standard histology and bioluminescence imaging (BLI). μCT enabled us to visualize diffuse tumor morphology and also to extract four different biomarkers that quantify not only tumor load but also aerated space in the lung as a marker of vital lung capacity and potential compensatory mechanisms. Complementary to BLI, applying this novel μCT-based approach enabled us to unravel sensitively and efficiently differences in metastatic potential between two cellular clones. In conclusion, μCT and BLI offer biomarkers that describe different and complementary aspects of lung metastasis, underlining the importance of multimodality follow-up. The added value of μCT findings is important to better assess lung metastasis and host/lung response in preclinical studies, which will be valuable for translational applications.
Collapse
|
14
|
Maji S, Chaudhary P, Akopova I, Nguyen PM, Hare RJ, Gryczynski I, Vishwanatha JK. Exosomal Annexin II Promotes Angiogenesis and Breast Cancer Metastasis. Mol Cancer Res 2016; 15:93-105. [PMID: 27760843 DOI: 10.1158/1541-7786.mcr-16-0163] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/05/2016] [Accepted: 10/03/2016] [Indexed: 12/18/2022]
Abstract
Tumor-derived exosomes are emerging mediators of tumorigenesis and tissue-specific metastasis. Proteomic profiling has identified Annexin II as one of the most highly expressed proteins in exosomes; however, studies focused on the biological role of exosomal Annexin II (exo-Anx II) are still lacking. In this study, mechanistic insight was sought regarding exo-Anx II and its function in angiogenesis and breast cancer metastasis. Multiple in vitro and in vivo techniques were used to study the role of exo-Anx II in angiogenesis. Using atomic force microscopy and Western blotting, exo-Anx II expression was characterized in normal and breast cancer cells. In addition, organ-specific metastatic breast cancer cells and animal models were used to define the role exo-Anx II in breast cancer metastasis. Results revealed that exo-Anx II expression is significantly higher in malignant cells than normal and premetastatic breast cancer cells. In vitro and in vivo studies demonstrated that exo-Anx II promotes tPA-dependent angiogenesis. Furthermore, in vivo analysis indicated that metastatic exosomes create a favorable microenvironment for metastasis, and exo-Anx II plays an important role in this process, as priming with Anx II-depleted exosomes reduces brain (∼4-fold) and lung (∼2-fold) metastasis. Upon delineating the mechanism, it was discovered that exo-Anx II causes macrophage-mediated activation of the p38MAPK, NF-κB, and STAT3 pathways and increased secretion of IL6 and TNFα. These data demonstrate an important role for exo-Anx II in breast cancer pathogenesis. IMPLICATIONS Exosome-associated Annexin II plays an important role in angiogenesis and breast cancer metastasis, which can be exploited as a potential biomarker as well as a therapeutic target for diagnosis and treatment of metastatic breast cancer. Mol Cancer Res; 15(1); 93-105. ©2016 AACR.
Collapse
Affiliation(s)
- Sayantan Maji
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas. .,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas
| | - Pankaj Chaudhary
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas.,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas
| | - Irina Akopova
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - Phung M Nguyen
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | | | - Ignacy Gryczynski
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
| | - Jamboor K Vishwanatha
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Fort Worth, Texas.,Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas.,Texas Center for Health Disparities, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
15
|
Iwanowycz S, Wang J, Hodge J, Wang Y, Yu F, Fan D. Emodin Inhibits Breast Cancer Growth by Blocking the Tumor-Promoting Feedforward Loop between Cancer Cells and Macrophages. Mol Cancer Ther 2016; 15:1931-42. [PMID: 27196773 DOI: 10.1158/1535-7163.mct-15-0987] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
Abstract
Macrophage infiltration correlates with severity in many types of cancer. Tumor cells recruit macrophages and educate them to adopt an M2-like phenotype through the secretion of chemokines and growth factors, such as MCP1 and CSF1. Macrophages in turn promote tumor growth through supporting angiogenesis, suppressing antitumor immunity, modulating extracellular matrix remodeling, and promoting tumor cell migration. Thus, tumor cells and macrophages interact to create a feedforward loop supporting tumor growth and metastasis. In this study, we tested the ability of emodin, a Chinese herb-derived compound, to inhibit breast cancer growth in mice and examined the underlying mechanisms. Emodin was used to treat mice bearing EO771 or 4T1 breast tumors. It was shown that emodin attenuated tumor growth by inhibiting macrophage infiltration and M2-like polarization, accompanied by increased T-cell activation and reduced angiogenesis in tumors. The tumor inhibitory effects of emodin were lost in tumor-bearing mice with macrophage depletion. Emodin inhibited IRF4, STAT6, and C/EBPβ signaling and increased inhibitory histone H3 lysine 27 tri-methylation (H3K27m3) on the promoters of M2-related genes in tumor-associated macrophages. In addition, emodin inhibited tumor cell secretion of MCP1 and CSF1, as well as expression of surface anchoring molecule Thy-1, thus suppressing macrophage migration toward and adhesion to tumor cells. These results suggest that emodin acts on both breast cancer cells and macrophages and effectively blocks the tumor-promoting feedforward loop between the two cell types, thereby inhibiting breast cancer growth and metastasis. Mol Cancer Ther; 15(8); 1931-42. ©2016 AACR.
Collapse
Affiliation(s)
- Stephen Iwanowycz
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Junfeng Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Johnie Hodge
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Fang Yu
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina. Department of Nutrition and Food Hygiene, Fourth Military Medical University, Xi'an, China
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina.
| |
Collapse
|
16
|
Jang SJ, Kang JH, Lee YJ, Kim KI, Lee TS, Choe JG, Lim SM. Detection of metastatic tumors after γ-irradiation using longitudinal molecular imaging and gene expression profiling of metastatic tumor nodules. Int J Oncol 2016; 48:1361-8. [PMID: 26892334 PMCID: PMC4777593 DOI: 10.3892/ijo.2016.3384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/11/2016] [Indexed: 12/26/2022] Open
Abstract
A few recent reports have indicated that metastatic growth of several human cancer cells could be promoted by radiotherapy. C6-L cells expressing the firefly luciferase (fLuc) gene were implanted subcutaneously into the right thigh of BALB/c nu/nu mice. C6-L xenograft mice were treated locally with 50-Gy γ-irradiation (γ-IR) in five 10-Gy fractions. Metastatic tumors were evaluated after γ-IR by imaging techniques. Total RNA from non-irradiated primary tumor (NRPT), γ-irradiated primary tumor (RPT), and three metastatic lung nodule was isolated and analyzed by microarray. Metastatic lung nodules were detected by BLI and PET/CT after 6–9 weeks of γ-IR in 6 (17.1%) of the 35 mice. The images clearly demonstrated high [18F]FLT and [18F]FDG uptake into metastatic lung nodules. Whole mRNA expression patterns were analyzed by microarray to elucidate the changes among NRPT, RPT and metastatic lung nodules after γ-IR. In particular, expression changes in the cancer stem cell markers were highly significant in RPT. We observed the metastatic tumors after γ-IR in a tumor-bearing animal model using molecular imaging methods and analyzed the gene expression profile to elucidate genetic changes after γ-IR.
Collapse
Affiliation(s)
- Su Jin Jang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Joo Hyun Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Yong Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Kwang Il Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Tae Sup Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| | - Jae Gol Choe
- Department of Nuclear Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Sang Moo Lim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 139-706, Republic of Korea
| |
Collapse
|
17
|
Gagnon KB, Caine S, Samadi N, Martinson M, van der Loop M, Alcorn J, Chapman LD, Belev G, Nichol H. Design of a mouse restraint for synchrotron-based computed tomography imaging. JOURNAL OF SYNCHROTRON RADIATION 2015; 22:1297-1300. [PMID: 26289283 DOI: 10.1107/s160057751501036x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/29/2015] [Indexed: 06/04/2023]
Abstract
High-resolution computed tomography (CT) imaging of a live animal within a lead-lined synchrotron light hutch presents several unique challenges. In order to confirm that the animal is under a stable plane of anaesthesia, several physiological parameters (e.g. heart rate, arterial oxygen saturation, core body temperature and respiratory rate) must be remotely monitored from outside the imaging hutch. In addition, to properly scan the thoracic region using CT, the animal needs to be held in a vertical position perpendicular to the fixed angle of the X-ray beam and free to rotate 180°-360°. A new X-ray transparent mouse restraint designed and fabricated using computer-aided design software and three-dimensional rapid prototype printing has been successfully tested at the Biomedical Imaging and Therapy bending-magnet (BMIT-BM) beamline at the Canadian Light Source.
Collapse
Affiliation(s)
- Kenneth B Gagnon
- Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sally Caine
- Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Nazanin Samadi
- Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mercedes Martinson
- Physics and Engineering Physics, University of Saskatchewan, Saskatoon, SK, Canada
| | - Melanie van der Loop
- Research Services and Ethics Office, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jane Alcorn
- Research Services and Ethics Office, University of Saskatchewan, Saskatoon, SK, Canada
| | - L Dean Chapman
- Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - George Belev
- Biomedical Imaging and Therapy Beamlines, Canadian Light Source, Saskatoon, SK, Canada
| | - Helen Nichol
- Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|