1
|
Milshteyn L, Villamejor A, Merchant A, Lownik J. A novel murine syngeneic CD8 peripheral T-cell lymphoma model with preclinical applications. Leuk Lymphoma 2024:1-7. [PMID: 39291652 DOI: 10.1080/10428194.2024.2404253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/26/2024] [Accepted: 09/08/2024] [Indexed: 09/19/2024]
Abstract
Peripheral T-cell Lymphoma (PTCL) represents a heterogenous group of aggressive non-Hodgkin Lymphomas with poor prognostic outcomes and limited treatment options. The development and refinement of therapeutic strategies for PTCL are impeded by a paucity of reliable preclinical models that accurately mimic the disease's pathophysiology. There is a dire need for more physiologically relevant models for PTCL. Here we describe a spontaneousCD8+ peripheral T-cell lymphoma cell line (LM-23) derived from a 12-week-old female Balb/cJ mouse. Both intravenous and subcutaneous administration of this cell line to syngeneic Balb/cJ mice resulted in rapid establishment of tumor growth. CHOP and anti-PD1 treatment both displayed no benefit to mice in regulating tumor growth. Such results along with its phenotypic characteristics, rapid growth, and metastatic behavior in syngeneic mice highlight its value in studying the elusive disease and discovery of novel therapeutics.
Collapse
Affiliation(s)
| | | | | | - Joseph Lownik
- Department of Pathology & Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
2
|
Fernandez-Gil BI, Schiapparelli P, Navarro-Garcia de Llano JP, Otamendi-Lopez A, Ulloa-Navas MJ, Michaelides L, Vazquez-Ramos CA, Herchko SM, Murray ME, Cherukuri Y, Asmann YW, Trifiletti DM, Quiñones-Hinojosa A. Effects of PreOperative radiotherapy in a preclinical glioblastoma model: a paradigm-shift approach. J Neurooncol 2024; 169:633-646. [PMID: 39037687 DOI: 10.1007/s11060-024-04765-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/29/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE PreOperative radiotherapy (RT) is commonly used in the treatment of brain metastasis and different cancer types but has never been used in primary glioblastoma (GBM). Here, we aim to establish, describe, and validate the use of PreOperative RT for the treatment of GBM in a preclinical model. METHODS Rat brains were locally irradiated with 30-Gy, hypofractionated in five doses 2 weeks before or after the resection of intracranial GBM. Kaplan-Meier analysis determined survival. Hematoxylin-eosin staining was performed, and nuclei size and p21 senescence marker were measured in both resected and recurrent rodent tumors. Immunohistochemistry assessed microglia/macrophage markers, and RNAseq analyzed gene expression changes in recurrent tumors. Akoya Multiplex Staining on two human patients from our ongoing Phase I/IIa trial served as proof of principle. RESULTS PreOperative RT group median survival was significantly higher than PostOperative RT (p < 0.05). Radiation enlarged cytoplasm and nuclei in PreOperative RT resected tumors (p < 0.001) and induced senescence in PostOperative RT recurrent tumors (p < 0.05). Gene Set Enrichment Analysis (GSEA) suggested a more proliferative profile in PreOperative RT group. PreOperative RT showed lower macrophage/microglia recruitment in recurrent tumors (p < 0.01) compared to PostOperative RT. Akoya Multiplex results indicated TGF-ß accumulation in the cytoplasm of TAMs and CD4 + lymphocyte predominance in PostOperative group. CONCLUSIONS This is the first preclinical study showing feasibility and longer overall survival using neoadjuvant radiotherapy before GBM resection in a mammalian model. This suggests strong superiority for new clinical radiation strategies. Further studies and trials are required to confirm our results.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Steven M Herchko
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Melissa E Murray
- Department of Molecular Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yesesri Cherukuri
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | | |
Collapse
|
3
|
Garate-Soraluze E, Marco-Sanz J, Serrano-Mendioroz I, Marrodán L, Fernandez-Rubio L, Labiano S, Rodríguez-Ruiz ME. Radiotherapy protocols for mouse cancer model. Methods Cell Biol 2024; 185:99-113. [PMID: 38556454 DOI: 10.1016/bs.mcb.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Radiotherapy is a crucial treatment modality for cancer patients, with approximately 60% of individuals undergoing ionizing radiation as part of their disease management. In recent years, there has been a growing trend toward minimizing irradiation fields through the use of image-guided dosimetry and innovative technologies. These advancements allow for selective irradiation, delivering higher local doses while reducing the number of treatment sessions. Consequently, computer-assisted methods have significantly enhanced the effectiveness of radiotherapy in the curative and palliative treatment of various cancers. Although radiation therapy alone can effectively achieve local control in some cancer types, it may not be sufficient for others. As a result, further preclinical research is necessary to explore novel approaches including new schedules of radiotherapy treatments. Unfortunately, there is a concerning lack of correlation between clinical outcomes and experiments conducted on mouse models. We hypothesize that this disparity arises from the differences in irradiation strategies employed in preclinical studies compared to those used in clinical practice, which ultimately affects the translatability of findings to patients. In this study, we present two comprehensive radiotherapy protocols for the treatment of orthotopic melanoma and glioblastoma tumors. These protocols utilize a small animal radiation research platform, which is an ideal radiation device for delivering localized and precise X-ray doses to the tumor mass. By employing these platforms, we aim to limit the side effects associated with irradiating healthy surrounding tissues. Our detailed protocols offer a valuable framework for conducting preclinical studies that closely mimic clinical radiotherapy techniques, bridging the gap between experimental results and patient outcomes.
Collapse
Affiliation(s)
- Eneko Garate-Soraluze
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Javier Marco-Sanz
- Program of Solid Tumors, Center for Applied Medical Research (CIMA), Pamplona, Spain; Department of Pediatrics, University of Navarra Clinic, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Irantzu Serrano-Mendioroz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Lucía Marrodán
- Program of Solid Tumors, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Leticia Fernandez-Rubio
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Sara Labiano
- Program of Solid Tumors, Center for Applied Medical Research (CIMA), Pamplona, Spain; Department of Pediatrics, University of Navarra Clinic, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - María E Rodríguez-Ruiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Department of Radiation Oncology, University of Navarra Clinic, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
4
|
Winters TA, Cassatt DR, Harrison-Peters JR, Hollingsworth BA, Rios CI, Satyamitra MM, Taliaferro LP, DiCarlo AL. Considerations of Medical Preparedness to Assess and Treat Various Populations During a Radiation Public Health Emergency. Radiat Res 2023; 199:301-318. [PMID: 36656560 PMCID: PMC10120400 DOI: 10.1667/rade-22-00148.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/21/2022] [Indexed: 01/20/2023]
Abstract
During a radiological or nuclear public health emergency, given the heterogeneity of civilian populations, it is incumbent on medical response planners to understand and prepare for a potentially high degree of interindividual variability in the biological effects of radiation exposure. A part of advanced planning should include a comprehensive approach, in which the range of possible human responses in relation to the type of radiation expected from an incident has been thoughtfully considered. Although there are several reports addressing the radiation response for special populations (as compared to the standard 18-45-year-old male), the current review surveys published literature to assess the level of consideration given to differences in acute radiation responses in certain sub-groups. The authors attempt to bring clarity to the complex nature of human biology in the context of radiation to facilitate a path forward for radiation medical countermeasure (MCM) development that may be appropriate and effective in special populations. Consequently, the focus is on the medical (as opposed to logistical) aspects of preparedness and response. Populations identified for consideration include obstetric, pediatric, geriatric, males, females, individuals of different race/ethnicity, and people with comorbidities. Relevant animal models, biomarkers of radiation injury, and MCMs are highlighted, in addition to underscoring gaps in knowledge and the need for consistent and early inclusion of these populations in research. The inclusion of special populations in preclinical and clinical studies is essential to address shortcomings and is an important consideration for radiation public health emergency response planning. Pursuing this goal will benefit the population at large by considering those at greatest risk of health consequences after a radiological or nuclear mass casualty incident.
Collapse
Affiliation(s)
- Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Jenna R. Harrison-Peters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
- Current address: Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, Maryland
| | - Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
5
|
Entezam A, Fielding A, Bradley D, Fontanarosa D. Absorbed dose calculation for a realistic CT-derived mouse phantom irradiated with a standard Cs-137 cell irradiator using a Monte Carlo method. PLoS One 2023; 18:e0280765. [PMID: 36730280 PMCID: PMC9928120 DOI: 10.1371/journal.pone.0280765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 01/07/2023] [Indexed: 02/03/2023] Open
Abstract
Computed tomography (CT) derived Monte Carlo (MC) phantoms allow dose determination within small animal models that is not feasible with in-vivo dosimetry. The aim of this study was to develop a CT-derived MC phantom generated from a mouse with a xenograft tumour that could then be used to calculate both the dose heterogeneity in the tumour volume and out of field scattered dose for pre-clinical small animal irradiation experiments. A BEAMnrc Monte-Carlo model has been built of our irradiation system that comprises a lead collimator with a 1 cm diameter aperture fitted to a Cs-137 gamma irradiator. The MC model of the irradiation system was validated by comparing the calculated dose results with dosimetric film measurement in a polymethyl methacrylate (PMMA) phantom using a 1D gamma-index analysis. Dose distributions in the MC mouse phantom were calculated and visualized on the CT-image data. Dose volume histograms (DVHs) were generated for the tumour and organs at risk (OARs). The effect of the xenographic tumour volume on the scattered out of field dose was also investigated. The defined gamma index analysis criteria were met, indicating that our MC simulation is a valid model for MC mouse phantom dose calculations. MC dose calculations showed a maximum out of field dose to the mouse of 7% of Dmax. Absorbed dose to the tumour varies in the range 60%-100% of Dmax. DVH analysis demonstrated that tumour received an inhomogeneous dose of 12 Gy-20 Gy (for 20 Gy prescribed dose) while out of field doses to all OARs were minimized (1.29 Gy-1.38 Gy). Variation of the xenographic tumour volume exhibited no significant effect on the out of field scattered dose to OARs. The CT derived MC mouse model presented here is a useful tool for tumour dose verifications as well as investigating the doses to normal tissue (in out of field) for preclinical radiobiological research.
Collapse
Affiliation(s)
- Amir Entezam
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
- * E-mail:
| | - Andrew Fielding
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - David Bradley
- Centre for Applied Physics and Radiation Technologies, Sunway University, PJ, Malaysia
- Department of Physics, University of Surrey, Guildford, United Kingdom
| | - Davide Fontanarosa
- School of Clinical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
6
|
Wang LM, Yadav R, Serban M, Arias O, Seuntjens J, Ybarra N. Validation of an orthotopic non-small cell lung cancer mouse model, with left or right tumor growths, to use in conformal radiotherapy studies. PLoS One 2023; 18:e0284282. [PMID: 37053154 PMCID: PMC10101527 DOI: 10.1371/journal.pone.0284282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Orthotopic non-small cell lung cancer (NSCLC) mice models are important for establishing translatability of in vitro results. However, most orthotopic lung models do not produce localized tumors treatable by conformal radiotherapy (RT). Here we report on the performance of an orthotopic mice model featuring conformal RT treatable tumors following either left or right lung tumor cell implantation. Athymic Nude mice were surgically implanted with H1299 NSCLC cell line in either the left or right lung. Tumor development was tracked bi-weekly using computed tomography (CT) imaging. When lesions reached an appropriate size for treatment, animals were separated into non-treatment (control group) and RT treated groups. Both RT treated left and right lung tumors which were given a single dose of 20 Gy of 225 kV X-rays. Left lung tumors were treated with a two-field parallel opposed plan while right lung tumors were treated with a more conformal four-field plan to assess tumor control. Mice were monitored for 30 days after RT or after tumor reached treatment size for non-treatment animals. Treatment images from the left and right lung tumor were also used to assess the dose distribution for four distinct treatment plans: 1) Two sets of perpendicularly staggered parallel opposed fields, 2) two fields positioned in the anterior-posterior and posterior-anterior configuration, 3) an 180° arc field from 0° to 180° and 4) two parallel opposed fields which cross through the contralateral lung. Tumor volumes and changes throughout the follow-up period were tracked by three different types of quantitative tumor size approximation and tumor volumes derived from contours. Ultimately, our model generated delineable and conformal RT treatable tumor following both left and right lung implantation. Similarly consistent tumor development was noted between left and right models. We were also able to demonstrate that a single 20 Gy dose of 225 kV X-rays applied to either the right or left lung tumor models had similar levels of tumor control resulting in similar adverse outcomes and survival. And finally, three-dimensional tumor approximation featuring volume computed from the measured length across three perpendicular axes gave the best approximation of tumor volume, most closely resembled tumor volumes obtained with contours.
Collapse
Affiliation(s)
- Li Ming Wang
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Cancer Research Program, Research Institute of the McGill University Healthcare Centre, Montreal, Quebec, Canada
| | - Ranjan Yadav
- Medical Physics Unit, Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Monica Serban
- Radiation Medicine, Princess Margaret Cancer Centre and Department of Radiation Oncology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Osvaldo Arias
- Cancer Research Program, Research Institute of the McGill University Healthcare Centre, Montreal, Quebec, Canada
| | - Jan Seuntjens
- Radiation Medicine, Princess Margaret Cancer Centre and Department of Radiation Oncology and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Oncology, McGill University, Montreal, Quebec, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Norma Ybarra
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Cancer Research Program, Research Institute of the McGill University Healthcare Centre, Montreal, Quebec, Canada
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Potiron V, Delpon G, Ollivier L, Vaugier L, Doré M, Guimas V, Rio E, Thillays F, Llagostera C, Moignier A, Josset S, Chiavassa S, Perennec T, Supiot S. [Clinical research in radiation oncology: how to move from the laboratory to the patient?]. Cancer Radiother 2022; 26:808-813. [PMID: 35999162 DOI: 10.1016/j.canrad.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Translational research in radiation oncology is undergoing intense development. An increasingly rapid transfer is taking place from the laboratory to the patients, both in the selection of patients who can benefit from radiotherapy and in the development of innovative irradiation strategies or the development of combinations with drugs. Accelerating the passage of discoveries from the laboratory to the clinic represents the ideal of any translational research program but requires taking into account the multiple obstacles that can slow this progress. The ambition of the RadioTransNet network, a project to structure preclinical research in radiation oncology in France, is precisely to promote scientific and clinical interactions at the interface of radiotherapy and radiobiology, in its preclinical positioning, in order to identify priorities for strategic research dedicated to innovation in radiotherapy. The multidisciplinary radiotherapy teams with experts in biology, medicine, medical physics, mathematics and engineering sciences are able to meet these new challenges which will allow these advances to be made available to patients as quickly as possible.
Collapse
Affiliation(s)
- V Potiron
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France; Unité en sciences biologiques et biotechnologies, UMR CNRS 6286, 2, rue de la Houssinière, 44322 Nantes, France
| | - G Delpon
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France; IMT Atlantique, UMR CNRS 6457/IN2P3, Subatech, laboratoire de physique subatomique et des technologies associées, Nantes, France
| | - L Ollivier
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - L Vaugier
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - M Doré
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - V Guimas
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - E Rio
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - F Thillays
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - C Llagostera
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - A Moignier
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - S Josset
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - S Chiavassa
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France; IMT Atlantique, UMR CNRS 6457/IN2P3, Subatech, laboratoire de physique subatomique et des technologies associées, Nantes, France
| | - T Perennec
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France
| | - S Supiot
- Institut de cancérologie de l'Ouest, boulevard Jacques-Monod, 44800 Saint-Herblain, France; Unité en sciences biologiques et biotechnologies, UMR CNRS 6286, 2, rue de la Houssinière, 44322 Nantes, France.
| |
Collapse
|
8
|
Milot MC, Benesty OB, Dumulon-Perreault V, Ait-Mohand S, Richard PO, Rousseau É, Guérin B. 64Cu-DOTHA 2-PSMA, a Novel PSMA PET Radiotracer for Prostate Cancer with a Long Imaging Time Window. Pharmaceuticals (Basel) 2022; 15:ph15080996. [PMID: 36015144 PMCID: PMC9412875 DOI: 10.3390/ph15080996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer imaging and late-stage management can be improved with prostate-specific membrane antigen (PSMA)-targeting radiotracers. We developed a PSMA positron emission tomography (PET) radiotracer, DOTHA2-PSMA radiolabeled with 64Cu (T1/2: 12.7 h), to leverage its large imaging time window. This preclinical study aimed to evaluate the biological and imaging properties of 64Cu-DOTHA2-PSMA. Its stability was assessed in plasma ex vivo and in mice. Cellular behavior was studied for up to 48 h in LNCaP cells. Biodistribution studies were performed in balb/c mice for up to 48 h. Dynamic (1 h) and static (4 h and 24 h) PET imaging was completed in LNCaP tumor-bearing mice. 64Cu-DOTHA2-PSMA was stable ex vivo in plasma and reached cellular internalization up to 34.1 ± 4.9% injected activity (IA)/106 cells at 48 h post-injection (p.i.). Biodistribution results showed significantly lower uptake in kidneys than 68Ga-PSMA-617, our reference PET tracer (p < 0.001), but higher liver uptake at 2 h p.i. (p < 0.001). PET images showed 64Cu-DOTHA2-PSMA’s highest tumoral uptake at 4 h p.i., with a significant difference between blocked and non-blocked groups from the time of injection to 24 h p.i. The high stability and tumor uptake with a long tumor imaging time window of 64Cu-DOTHA2-PSMA potentially contribute to the prostate cancer theranostic approach and its local recurrence detection.
Collapse
Affiliation(s)
- Marie-Christine Milot
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Ophélie Bélissant Benesty
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Véronique Dumulon-Perreault
- Sherbrooke Molecular Imaging Center (CIMS), Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), 3001, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Samia Ait-Mohand
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Patrick O. Richard
- Department of Surgery, Division of Urology, Faculty of Medicine and Health Sciences, University de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Étienne Rousseau
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Sherbrooke Molecular Imaging Center (CIMS), Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), 3001, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Sherbrooke Molecular Imaging Center (CIMS), Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), 3001, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
9
|
A High-Throughput In Vitro Radiobiology Platform for Megavoltage Photon Linear Accelerator Studies. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We designed and developed a multiwell tissue culture plate irradiation setup, and intensity modulated radiotherapy plans were generated for 96-, 24-, and 6-well tissue culture plates. We demonstrated concordance between planned and measured/imaged radiation dose profiles using radiochromic film, a 2D ion chamber array, and an electronic portal-imaging device. Cell viability, clonogenic potential, and g-H2AX foci analyses showed no significant differences between intensity-modulated radiotherapy and open-field, homogeneous irradiations. This novel platform may help to expedite radiobiology experiments within a clinical environment and may be used for wide-ranging ex vivo radiobiology applications.
Collapse
|
10
|
Parsels LA, Zhang Q, Karnak D, Parsels JD, Lam K, Willers H, Green MD, Rehemtulla A, Lawrence TS, Morgan MA. Translation of DNA Damage Response Inhibitors as Chemoradiation Sensitizers From the Laboratory to the Clinic. Int J Radiat Oncol Biol Phys 2021; 111:e38-e53. [PMID: 34348175 PMCID: PMC8602768 DOI: 10.1016/j.ijrobp.2021.07.1708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022]
Abstract
Combination therapies with agents targeting the DNA damage response (DDR) offer an opportunity to selectively enhance the therapeutic index of chemoradiation or eliminate use of chemotherapy altogether. The successful translation of DDR inhibitors to clinical use requires investigating both their direct actions as (chemo)radiosensitizers and their potential to stimulate tumor immunogenicity. Beginning with high-throughput screening using both viability and DNA damage-reporter assays, followed by validation in gold-standard radiation colony-forming assays and in vitro assessment of mechanistic effects on the DDR, we describe proven strategies and methods leading to the clinical development of DDR inhibitors both with radiation alone and in combination with chemoradiation. Beyond these in vitro studies, we discuss the impact of key features of human xenograft and syngeneic mouse models on the relevance of in vivo tumor efficacy studies, particularly with regard to the immunogenic effects of combined therapy with radiation and DDR inhibitors. Finally, we describe recent technological advances in radiation delivery (using the small animal radiation research platform) that allow for conformal, clinically relevant radiation therapy in mouse models. This overall approach is critical to the successful clinical development and ultimate Food and Drug Administration approval of DDR inhibitors as (chemo)radiation sensitizers.
Collapse
Affiliation(s)
- Leslie A Parsels
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Qiang Zhang
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - David Karnak
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Joshua D Parsels
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Kwok Lam
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael D Green
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan.
| |
Collapse
|
11
|
Singh J, Hatcher S, Ku AA, Ding Z, Feng FY, Sharma RA, Pfister SX. Model Selection for the Preclinical Development of New Drug-Radiotherapy Combinations. Clin Oncol (R Coll Radiol) 2021; 33:694-704. [PMID: 34474951 DOI: 10.1016/j.clon.2021.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/13/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022]
Abstract
Radiotherapy plays an essential role in the treatment of more than half of all patients with cancer. In recent decades, advances in devices that deliver radiation and the development of treatment planning software have helped radiotherapy attain precise tumour targeting with minimal toxicity to surrounding tissues. Simultaneously, as more targeted drug therapies are being brought into the market, there has been significant interest in improving cure rates for cancer by adding drugs to radiotherapy to widen the therapeutic window, the difference between normal tissue toxicity and treatment efficacy. The development of new combination therapies will require judicious adaptation of preclinical models that are routinely used for traditional drug discovery. Here we highlight the strengths and weaknesses of each of these preclinical models and discuss how they can be used optimally to identify new and clinically beneficial drug-radiotherapy combinations.
Collapse
Affiliation(s)
- J Singh
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA
| | - S Hatcher
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA
| | - A A Ku
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA
| | - Z Ding
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA
| | - F Y Feng
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA; Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, California, USA; Department of Radiation Oncology, University of California, San Francisco, California, USA; Department of Urology, University of California, San Francisco, California, USA
| | - R A Sharma
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA; UCL Cancer Institute, University College London, London, UK
| | - S X Pfister
- Global Translational Science, Varian, a Siemens Healthineers company, Palo Alto, California, USA.
| |
Collapse
|
12
|
Willers H, Pan X, Borgeaud N, Korovina I, Koi L, Egan R, Greninger P, Rosenkranz A, Kung J, Liss AS, Parsels LA, Morgan MA, Lawrence TS, Lin SH, Hong TS, Yeap BY, Wirth L, Hata AN, Ott CJ, Benes CH, Baumann M, Krause M. Screening and Validation of Molecular Targeted Radiosensitizers. Int J Radiat Oncol Biol Phys 2021; 111:e63-e74. [PMID: 34343607 DOI: 10.1016/j.ijrobp.2021.07.1694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 11/16/2022]
Abstract
The development of molecular targeted drugs with radiation and chemotherapy are critically important for improving the outcomes of patients with hard-to-treat, potentially curable cancers. However, too many preclinical studies have not translated into successful radiation oncology trials. Major contributing factors to this insufficiency include poor reproducibility of preclinical data, inadequate preclinical modeling of inter-tumoral genomic heterogeneity that influences treatment sensitivity in the clinic, and a reliance on tumor growth delay instead of local control (TCD50) endpoints. There exists an urgent need to overcome these barriers to facilitate successful clinical translation of targeted radiosensitizers. To this end, we have employed 3D cell culture assays to better model tumor behavior in vivo. Examples of successful prediction of in vivo effects with these 3D assays include radiosensitization of head and neck cancers by inhibiting epidermal growth factor receptor or focal adhesion kinase signaling, and radioresistance associated with oncogenic mutation of KRAS. To address the issue of tumor heterogeneity we leveraged institutional resources that allow high-throughput 3D screening of radiation combinations with small molecule inhibitors across genomically characterized cell lines from lung, head and neck, and pancreatic cancers. This high-throughput screen is expected to uncover genomic biomarkers that will inform the successful clinical translation of targeted agents from the NCI CTEP portfolio and other sources. Screening "hits" need to be subjected to refinement studies that include clonogenic assays, addition of disease-specific chemotherapeutics, target/biomarker validation, and integration of patient-derived tumor models. The chemoradiosensitizing activities of the most promising drugs should be confirmed in TCD50 assays in xenograft models with/without relevant biomarker and utilizing clinically relevant radiation fractionation. We predict that appropriately validated and biomarker-directed targeted therapies will have a higher likelihood than past efforts to be successfully incorporated into the standard management of hard-to-treat tumors.
Collapse
Affiliation(s)
- Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Xiao Pan
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nathalie Borgeaud
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), partner site Dresden
| | - Irina Korovina
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), partner site Dresden; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Regina Egan
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Patricia Greninger
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Aliza Rosenkranz
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jong Kung
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew S Liss
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leslie A Parsels
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Beow Y Yeap
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lori Wirth
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aaron N Hata
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Christopher J Ott
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Core center Heidelberg, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), partner site Dresden; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; National Center for Tumour Diseases (NCT), Partner site Dresden, Germany
| |
Collapse
|
13
|
Zhou Z, Zhu J, Jiang M, Sang L, Hao K, He H. The Combination of Cell Cultured Technology and In Silico Model to Inform the Drug Development. Pharmaceutics 2021; 13:pharmaceutics13050704. [PMID: 34065907 PMCID: PMC8151315 DOI: 10.3390/pharmaceutics13050704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Human-derived in vitro models can provide high-throughput efficacy and toxicity data without a species gap in drug development. Challenges are still encountered regarding the full utilisation of massive data in clinical settings. The lack of translated methods hinders the reliable prediction of clinical outcomes. Therefore, in this study, in silico models were proposed to tackle these obstacles from in vitro to in vivo translation, and the current major cell culture methods were introduced, such as human-induced pluripotent stem cells (hiPSCs), 3D cells, organoids, and microphysiological systems (MPS). Furthermore, the role and applications of several in silico models were summarised, including the physiologically based pharmacokinetic model (PBPK), pharmacokinetic/pharmacodynamic model (PK/PD), quantitative systems pharmacology model (QSP), and virtual clinical trials. These credible translation cases will provide templates for subsequent in vitro to in vivo translation. We believe that synergising high-quality in vitro data with existing models can better guide drug development and clinical use.
Collapse
Affiliation(s)
- Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Jinwei Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Muhan Jiang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
- Correspondence: (K.H.); (H.H.)
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
- Correspondence: (K.H.); (H.H.)
| |
Collapse
|
14
|
Wang L, Wu J, Chen J, Dou W, Zhao Q, Han J, Liu J, Su W, Li A, Liu P, An Z, Xu C, Sun Y. Advances in reconstructing intestinal functionalities in vitro: From two/three dimensional-cell culture platforms to human intestine-on-a-chip. Talanta 2021; 226:122097. [PMID: 33676654 DOI: 10.1016/j.talanta.2021.122097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 12/20/2022]
Abstract
Standard two/three dimensional (2D/3D)-cell culture platforms have facilitated the understanding of the communications between various cell types and their microenvironments. However, they are still limited in recapitulating the complex functionalities in vivo, such as tissue formation, tissue-tissue interface, and mechanical/biochemical microenvironments of tissues and organs. Intestine-on-a-chip platforms offer a new way to mimic intestinal behaviors and functionalities by constructing in vitro intestinal models in microfluidic devices. This review summarizes the advances and limitations of the state-of-the-art 2D/3D-cell culture platforms, animal models, intestine chips, and the combined multi-organ chips related with intestines. Their applications to studying intestinal functions, drug testing, and disease modeling are introduced. Different intestinal cell sources are compared in terms of gene expression abilities and the recapitulated intestinal morphologies. Among these cells, cells isolated form human intestinal tissues and derived from pluripotent stem cells appear to be more suitable for in vitro reconstruction of intestinal organs. Key challenges of current intestine-on-a-chip platforms and future directions are also discussed.
Collapse
Affiliation(s)
- Li Wang
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jian Wu
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jun Chen
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Rd, Toronto, Ontario, M5S 3G8, Canada
| | - Qili Zhao
- Institute of Robotics and Automatic Information System (IRAIS) and the Tianjin Key Laboratory of Intelligent Robotic (tjKLIR), Nankai University, Tianjin, 300350, China
| | - Junlei Han
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jinliang Liu
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Weiguang Su
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Anqing Li
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Pengbo Liu
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Zhao An
- Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chonghai Xu
- Advanced Micro and Nano-instruments Center, School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Rd, Toronto, Ontario, M5S 3G8, Canada
| |
Collapse
|
15
|
Redler G, Pearson E, Liu X, Gertsenshteyn I, Epel B, Pelizzari C, Aydogan B, Weichselbaum R, Halpern HJ, Wiersma RD. Small Animal IMRT Using 3D-Printed Compensators. Int J Radiat Oncol Biol Phys 2020; 110:551-565. [PMID: 33373659 DOI: 10.1016/j.ijrobp.2020.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 11/17/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Preclinical radiation replicating clinical intensity modulated radiation therapy (IMRT) techniques can provide data translatable to clinical practice. For this work, treatment plans were created for oxygen-guided dose-painting in small animals using inverse-planned IMRT. Spatially varying beam intensities were achieved using 3-dimensional (3D)-printed compensators. METHODS AND MATERIALS Optimized beam fluence from arbitrary gantry angles was determined using a verified model of the XRAD225Cx treatment beam. Compensators were 3D-printed with varied thickness to provide desired attenuation using copper/polylactic-acid. Spatial resolution capabilities were investigated using printed test-patterns. Following American Association of Physicists in Medicine TG119, a 5-beam IMRT plan was created for a miniaturized (∼1/8th scale) C-shape target. Electron paramagnetic resonance imaging of murine tumor oxygenation guided simultaneous integrated boost (SIB) plans conformally treating tumor to a base dose (Rx1) with boost (Rx2) based on tumor oxygenation. The 3D-printed compensator intensity modulation accuracy and precision was evaluated by individually delivering each field to a phantom containing radiochromic film and subsequent per-field gamma analysis. The methodology was validated end-to-end with composite delivery (incorporating 3D-printed tungsten/polylactic-acid beam trimmers to reduce out-of-field leakage) of the oxygen-guided SIB plan to a phantom containing film and subsequent gamma analysis. RESULTS Resolution test-patterns demonstrate practical printer resolution of ∼0.7 mm, corresponding to 1.0 mm bixels at the isocenter. The miniaturized C-shape plan provides planning target volume coverage (V95% = 95%) with organ sparing (organs at risk Dmax < 50%). The SIB plan to hypoxic tumor demonstrates the utility of this approach (hypoxic tumor V95%,Rx2 = 91.6%, normoxic tumor V95%,Rx1 = 95.7%, normal tissue V100%,Rx1 = 7.1%). The more challenging SIB plan to boost the normoxic tumor rim achieved normoxic tumor V95%,Rx2 = 90.9%, hypoxic tumor V95%,Rx1 = 62.7%, and normal tissue V100%,Rx2 = 5.3%. Average per-field gamma passing rates using 3%/1.0 mm, 3%/0.7 mm, and 3%/0.5 mm criteria were 98.8% ± 2.8%, 96.6% ± 4.1%, and 90.6% ± 5.9%, respectively. Composite delivery of the hypoxia boost plan and gamma analysis (3%/1 mm) gave passing results of 95.3% and 98.1% for the 2 measured orthogonal dose planes. CONCLUSIONS This simple and cost-effective approach using 3D-printed compensators for small-animal IMRT provides a methodology enabling preclinical studies that can be readily translated into the clinic. The presented oxygen-guided dose-painting demonstrates that this methodology will facilitate studies driving much needed biologic personalization of radiation therapy for improvements in patient outcomes.
Collapse
Affiliation(s)
- Gage Redler
- Moffitt Cancer Center, Department of Radiation Oncology, Tampa, Florida.
| | - Erik Pearson
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Xinmin Liu
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Inna Gertsenshteyn
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Boris Epel
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Charles Pelizzari
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Bulent Aydogan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Howard J Halpern
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Rodney D Wiersma
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Yoon SW, Kodra J, Miles DA, Kirsch DG, Oldham M. A method for generating intensity-modulated radiation therapy fields for small animal irradiators utilizing 3D-printed compensator molds. Med Phys 2020; 47:4363-4371. [PMID: 32281657 DOI: 10.1002/mp.14175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/24/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the feasibility of using fused deposition modeling (FDM) three-dimensional (3D) printer to generate radiation compensators for high-resolution (~1 mm) intensity-modulated radiation therapy (IMRT) for small animal radiation treatment. We propose a novel method incorporating 3D-printed compensator molds filled with NaI powder. METHODS The inverse planning module of the computational environment for radiotherapy research (CERR) software was adapted to simulate the XRAD-225Cx irradiator, both geometry and kV beam quality (the latter using a phase space file provided for XRAD-225Cx). A nine-field IMRT treatment was created for a scaled-down version of the imaging and radiation oncology core (IROC) Head and Neck IMRT credentialing test, recreated on a 2.2-cm-diameter cylindrical phantom. Optimized fluence maps comprising nine fields and a total of 2564 beamlets were calculated at resolution of 1.25 × 1.25 mm2 . A hollow compensator mold was created (using in-house software and algorithm) for each field using 3D printing with polylactic acid (PLA) filaments. The molds were then packed with sodium iodide powder (NaI, measured density ρNaI = 2.062 g/cm3 ). The mounted compensator mold thickness was limited to 13.8 mm due to clearance issues with couch collision. At treatment delivery, each compensator was manually mounted to a customized block tray attached to the reference 40 × 40 mm2 collimator. Compensator reproducibility among three repeated 3D-printed molds was measured with Radiochromic EBT2 film. The two-dimensional (2D) dose distributions of the nine fields were compared to calculated 2D doses from CERR using gamma comparisons with distance-to-agreement criteria of 0.5-0.25 mm and dose difference criteria of 3-5%. RESULTS Good reproducibility of 3D-printed compensator manufacture was observed with mean error of ±0.024 Gy and relative dose error of ±4.2% within the modulated part of the beam. Within the limit of 13.8 mm compensator height, a maximum radiation blocking efficiency of 91.5% was achieved. Per field, about 45.5 g of NaI powder was used. Gamma analysis on each of the nine delivered IMRT fields using radiochromic films resulted in eight of nine treatment fields with >90% pass rate with 5%/0.5 mm tolerances. However, low gamma passing rate of 49-66% (3%/0.25 mm to 5%/0.5 mm) was noted in one field, attributed to fabrication errors resulting in over-filling the mold. The nine-field treatment plan was delivered in under 30 min with no mechanical or collisional issues. CONCLUSIONS We show the feasibility of high spatial resolution IMRT treatment on a small animal irradiator utilizing 3D-printed compensator shells packed with NaI powder. Using the PLA mold with NaI powder was attractive due to the ease of 3D printing a PLA mold at high geometric resolution and the well-balanced attenuation properties of NaI powders that prevented the mold from becoming too bulky. IMRT fields with 1.25-mm resolution are capable with significant fluence modulation with relative dose accuracy of ±4.2%.
Collapse
Affiliation(s)
- Suk W Yoon
- Department of Radiation Oncology, Perelman School of Medicine of the University of Pennsylvania, Perelman Center for Advanced Medicine, Philadelphia, PA, 19104, USA.,Medical Physics Graduate Program, Duke University, Durham, NC, 27705, USA
| | - Jacob Kodra
- Medical Physics Graduate Program, Duke University, Durham, NC, 27705, USA
| | - Devin A Miles
- UW School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27705, USA
| | - Mark Oldham
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27705, USA
| |
Collapse
|
17
|
Chargari C, Levy A, Paoletti X, Soria JC, Massard C, Weichselbaum RR, Deutsch E. Methodological Development of Combination Drug and Radiotherapy in Basic and Clinical Research. Clin Cancer Res 2020; 26:4723-4736. [PMID: 32409306 DOI: 10.1158/1078-0432.ccr-19-4155] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/14/2020] [Accepted: 05/12/2020] [Indexed: 01/03/2023]
Abstract
Newer technical improvements in radiation oncology have been rapidly implemented in recent decades, allowing an improved therapeutic ratio. The development of strategies using local and systemic treatments concurrently, mainly targeted therapies, has however plateaued. Targeted molecular compounds and immunotherapy are increasingly being incorporated as the new standard of care for a wide array of cancers. A better understanding of possible prior methodology issues is therefore required and should be integrated into upcoming early clinical trials including individualized radiotherapy-drug combinations. The outcome of clinical trials is influenced by the validity of the preclinical proofs of concept, the impact on normal tissue, the robustness of biomarkers and the quality of the delivery of radiation. Herein, key methodological aspects are discussed with the aim of optimizing the design and implementation of future precision drug-radiotherapy trials.
Collapse
Affiliation(s)
- Cyrus Chargari
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Institut de Recherche Biomédicale des Armées, Brétigny sur Orge, France
| | - Antonin Levy
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Xavier Paoletti
- University of Versailles St. Quentin, France
- Institut Curie INSERM U900, Biostatistics for Personalized Medicine Team, St. Cloud, France
| | | | - Christophe Massard
- Université Paris-Sud, Orsay, France
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
- Université Paris-Sud, Orsay, France
- INSERM U1030, Molecular Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
18
|
Contreras HR, López-Moncada F, Castellón EA. Cancer stem cell and mesenchymal cell cooperative actions in metastasis progression and hormone resistance in prostate cancer: Potential role of androgen and gonadotropin‑releasing hormone receptors (Review). Int J Oncol 2020; 56:1075-1082. [PMID: 32319606 DOI: 10.3892/ijo.2020.5008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of male cancer‑associated mortality worldwide. Mortality is associated with metastasis and hormone resistance. Cellular, genetic and molecular mechanisms underlying metastatic progression and hormone resistance are poorly understood. Studies have investigated the local effects of gonadotropin‑releasing hormone (GnRH) analogs (used for androgen deprivation treatments) and the presence of the GnRH receptor (GnRH‑R) on PCa cells. Furthermore, cell subpopulations with stem‑like properties, or cancer stem cells, have been isolated and characterized using a cell culture system derived from explants of human prostate tumors. In addition, the development of preclinical orthotopic models of human PCa in a nonobese diabetic/severe combined immunodeficiency mouse model of compromised immunity has enabled the establishment of a reproducible system of metastatic progression in vivo. There is increasing evidence that metastasis is a complex process involving the cooperative actions of different cancer cell subpopulations, in which cancer stem‑like cells would be responsible for the final step of colonizing premetastatic niches. It has been hypothesized that PCa cells with stemness and mesenchymal signatures act cooperatively in metastatic progression and the inhibition of stemness genes, and that overexpression of androgen receptor (AR) and GnRH‑R decreases the rate the metastasis and sensitizes tumors to hormone therapy. The aim of the present review is to analyze the evidence regarding this cooperative process and the possible influence of stem‑like cell phenotypes, AR and GnRH‑R in metastatic progression and hormone resistance. These aspects may represent an important contribution in the understanding of the mechanisms underlying metastasis and hormone resistance in PCa, and potential routes to blocking these processes, enabling the development of novel therapies that would be particularly relevant for patients with metastatic and castration‑resistant PCa.
Collapse
Affiliation(s)
- Héctor R Contreras
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Fernanda López-Moncada
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Enrique A Castellón
- Laboratory of Cellular and Molecular Oncology, Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
19
|
Cosper PF, Abel L, Lee YS, Paz C, Kaushik S, Nickel KP, Alexandridis R, Scott JG, Bruce JY, Kimple RJ. Patient Derived Models to Study Head and Neck Cancer Radiation Response. Cancers (Basel) 2020; 12:E419. [PMID: 32059418 PMCID: PMC7072508 DOI: 10.3390/cancers12020419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/08/2020] [Accepted: 02/09/2020] [Indexed: 01/23/2023] Open
Abstract
Patient-derived model systems are important tools for studying novel anti-cancer therapies. Patient-derived xenografts (PDXs) have gained favor over the last 10 years as newer mouse strains have improved the success rate of establishing PDXs from patient biopsies. PDXs can be engrafted from head and neck cancer (HNC) samples across a wide range of cancer stages, retain the genetic features of their human source, and can be treated with both chemotherapy and radiation, allowing for clinically relevant studies. Not only do PDXs allow for the study of patient tissues in an in vivo model, they can also provide a renewable source of cancer cells for organoid cultures. Herein, we review the uses of HNC patient-derived models for radiation research, including approaches to establishing both orthotopic and heterotopic PDXs, approaches and potential pitfalls to delivering chemotherapy and radiation to these animal models, biological advantages and limitations, and alternatives to animal studies that still use patient-derived tissues.
Collapse
Affiliation(s)
- Pippa F. Cosper
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Lindsey Abel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Yong-Syu Lee
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Cristina Paz
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Saakshi Kaushik
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Kwangok P. Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (P.F.C.); (L.A.); (Y.-S.L.); (C.P.); (S.K.); (K.P.N.)
| | - Roxana Alexandridis
- Department of Biostatistics and Medical Informatics, UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Jacob G. Scott
- Departments of Translational Hematology and Oncology Research and Radiation Oncology, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Justine Y. Bruce
- Department of Medicine, Division of Hematology and Oncology, UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Randall J. Kimple
- Department of Human Oncology, UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
20
|
Black M, Ghasemi F, Sun RX, Stecho W, Datti A, Meens J, Pinto N, Ruicci KM, Khan MI, Han MW, Shaikh M, Yoo J, Fung K, MacNeil D, Palma DA, Winquist E, Howlett CJ, Mymryk JS, Ailles L, Boutros PC, Barrett JW, Nichols AC. Spleen tyrosine kinase expression is correlated with human papillomavirus in head and neck cancer. Oral Oncol 2019; 101:104529. [PMID: 31864959 DOI: 10.1016/j.oraloncology.2019.104529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 11/25/2019] [Accepted: 12/15/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Spleen tyrosine kinase (SYK) is a promoter of cell survival in a variety of cell types, including normal and cancerous epithelial cells. We hypothesized that SYK would an important therapeutic target to inhibit for the treatment of HNSCC. MATERIALS AND METHODS SYK protein abundance in patient tumours was evaluated. SYK protein and mRNA abundance was used to examine patient survival and human papillomavirus (HPV) status. Small-interfering RNAs and gene editing with CRISPR/Cas9 were used to evaluate SYK expression on proliferation in HNSCC cell lines. The potency of SYK inhibitor ER27319 maleate on cellular proliferation was tested using a panel of 28 HNSCC cell lines and in vivo in HNSCC patient-derived xenograft (PDX) models. RESULTS Moderate to high protein expression of SYK was observed in 24% of patient tumors and high SYK expression was exclusively observed in HPV-positive samples (p < 0.001). SYK inhibition with RNA interference, gene editing or a SYK inhibitor (ER27319) decreased cell proliferation and migration. Treatment of PDXs with ER27319 maleate was observed to reduce tumour burden in vivo in two of three models. CONCLUSIONS HPV-positive HNSCC harbours high SYK protein levels. We demonstrate that proliferation, migration and overall burden of these tumours can be reduced by genetic or pharmacologic inhibition of SYK. Taken together, these data establish SYK as a therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Morgan Black
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Farhad Ghasemi
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada
| | - Ren X Sun
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - William Stecho
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Alessandro Datti
- Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy.
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nicole Pinto
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Kara M Ruicci
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - M Imran Khan
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada
| | - Myung Woul Han
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada
| | - Mushfiq Shaikh
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada
| | - John Yoo
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Kevin Fung
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Danielle MacNeil
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - David A Palma
- Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Eric Winquist
- Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Christopher J Howlett
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Joe S Mymryk
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada; Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Paul C Boutros
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology, London Health Sciences Centre, London, Ontario, Canada; Department of Oncology, London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|
21
|
Woods K, Nguyen D, Neph R, Ruan D, O'Connor D, Sheng K. A sparse orthogonal collimator for small animal intensity-modulated radiation therapy part I: Planning system development and commissioning. Med Phys 2019; 46:5703-5713. [PMID: 31621920 DOI: 10.1002/mp.13872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To achieve more translatable preclinical research results, small animal irradiation needs to more closely simulate human radiotherapy. Although the clinical gold standard is intensity-modulated radiation therapy (IMRT), the direct translation of this method for small animals is impractical. In this study we describe the treatment planning system for a novel dose modulation device to address this challenge. METHODS Using delineated target and avoidance structures, a rectangular aperture optimization (RAO) problem was formulated to penalize deviations from a desired dose distribution and limit the number of selected rectangular apertures. RAO was used to create IMRT plans with highly concave targets in the mouse brain, and the plan quality was compared to that using a hypothetical miniaturized multileaf collimator (MLC). RAO plans were also created for a realistic application of mouse whole liver irradiation and for a highly complex two-dimensional (2D) dose distribution as a proof-of-principle. Beam commissioning data, including output and off-axis factors and percent depth dose (PDD) curves, were acquired for our small animal irradiation system and incorporated into the treatment planning system. A plan post-processing step was implemented for aperture size-specific dose recalculation and aperture weighting reoptimization. RESULTS The first RAO test case achieved highly conformal doses to concave targets in the brain, with substantially better dose gradient, conformity, and target dose homogeneity than the hypothetical miniaturized MLC plans. In the second test case, a highly conformal dose to the liver was achieved with significant sparing of the kidneys. RAO also successfully replicated a complex 2D dose distribution with three prescription dose levels. Energy spectra for field sizes 1 to 20 mm were calculated to match the measured PDD curves, with maximum and mean dose deviations of 4.47 ± 0.30% and 1.71 ± 0.18%. The final reoptimization of aperture weightings for the complex RAO test plan was able to reduce the maximum and mean dose deviations between the optimized and recalculated dose distributions from 10.3% to 6.6% and 4.0% to 2.8%, respectively. CONCLUSIONS Using the advanced optimization techniques, complex IMRT plans were achieved using a simple dose modulation device. Beam commissioning data were incorporated into the treatment planning process to more accurately predict the resulting dose distribution. This platform substantially reduces the gap in treatment plan quality between clinical and preclinical radiotherapy, potentially increasing the value and flexibility of small animal studies.
Collapse
Affiliation(s)
- Kaley Woods
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dan Nguyen
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ryan Neph
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dan Ruan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Daniel O'Connor
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ke Sheng
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
22
|
Dünker N, Jendrossek V. Implementation of the Chick Chorioallantoic Membrane (CAM) Model in Radiation Biology and Experimental Radiation Oncology Research. Cancers (Basel) 2019; 11:cancers11101499. [PMID: 31591362 PMCID: PMC6826367 DOI: 10.3390/cancers11101499] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) is part of standard cancer treatment. Innovations in treatment planning and increased precision in dose delivery have significantly improved the therapeutic gain of radiotherapy but are reaching their limits due to biologic constraints. Thus, a better understanding of the complex local and systemic responses to RT and of the biological mechanisms causing treatment success or failure is required if we aim to define novel targets for biological therapy optimization. Moreover, optimal treatment schedules and prognostic biomarkers have to be defined for assigning patients to the best treatment option. The complexity of the tumor environment and of the radiation response requires extensive in vivo experiments for the validation of such treatments. So far in vivo investigations have mostly been performed in time- and cost-intensive murine models. Here we propose the implementation of the chick chorioallantoic membrane (CAM) model as a fast, cost-efficient model for semi high-throughput preclinical in vivo screening of the modulation of the radiation effects by molecularly targeted drugs. This review provides a comprehensive overview on the application spectrum, advantages and limitations of the CAM assay and summarizes current knowledge of its applicability for cancer research with special focus on research in radiation biology and experimental radiation oncology.
Collapse
Affiliation(s)
- Nicole Dünker
- Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Medicine Essen, 45122 Essen, Germany.
| |
Collapse
|
23
|
Cardilin T, Almquist J, Jirstrand M, Zimmermann A, Lignet F, El Bawab S, Gabrielsson J. Modeling long-term tumor growth and kill after combinations of radiation and radiosensitizing agents. Cancer Chemother Pharmacol 2019; 83:1159-1173. [PMID: 30976845 PMCID: PMC6499765 DOI: 10.1007/s00280-019-03829-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/01/2019] [Indexed: 11/30/2022]
Abstract
PURPOSE Radiation therapy, whether given alone or in combination with chemical agents, is one of the cornerstones of oncology. We develop a quantitative model that describes tumor growth during and after treatment with radiation and radiosensitizing agents. The model also describes long-term treatment effects including tumor regrowth and eradication. METHODS We challenge the model with data from a xenograft study using a clinically relevant administration schedule and use a mixed-effects approach for model-fitting. We use the calibrated model to predict exposure combinations that result in tumor eradication using Tumor Static Exposure (TSE). RESULTS The model is able to adequately describe data from all treatment groups, with the parameter estimates taking biologically reasonable values. Using TSE, we predict the total radiation dose necessary for tumor eradication to be 110 Gy, which is reduced to 80 or 30 Gy with co-administration of 25 or 100 mg kg-1 of a radiosensitizer. TSE is also explored via a heat map of different growth and shrinkage rates. Finally, we discuss the translational potential of the model and TSE concept to humans. CONCLUSIONS The new model is capable of describing different tumor dynamics including tumor eradication and tumor regrowth with different rates, and can be calibrated using data from standard xenograft experiments. TSE and related concepts can be used to predict tumor shrinkage and eradication, and have the potential to guide new experiments and support translations from animals to humans.
Collapse
Affiliation(s)
- Tim Cardilin
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden. .,Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden.
| | - Joachim Almquist
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, Gothenburg, Sweden
| | - Astrid Zimmermann
- Translation Innovation Platform Oncology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Floriane Lignet
- Translational Medicine, Quantitative Pharmacology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Samer El Bawab
- Translational Medicine, Quantitative Pharmacology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Johan Gabrielsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
24
|
DuRoss AN, Neufeld MJ, Rana S, Thomas CR, Sun C. Integrating nanomedicine into clinical radiotherapy regimens. Adv Drug Deliv Rev 2019; 144:35-56. [PMID: 31279729 PMCID: PMC6745263 DOI: 10.1016/j.addr.2019.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 01/06/2023]
Abstract
While the advancement of clinical radiotherapy was driven by technological innovations throughout the 20th century, continued improvement relies on rational combination therapies derived from biological insights. In this review, we highlight the importance of combination radiotherapy in the era of precision medicine. Specifically, we survey and summarize the areas of research where improved understanding in cancer biology will propel the field of radiotherapy forward by allowing integration of novel nanotechnology-based treatments.
Collapse
Affiliation(s)
- Allison N DuRoss
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Megan J Neufeld
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Shushan Rana
- Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Charles R Thomas
- Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Radiation Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
25
|
Evolution of the Supermodel: Progress in Modelling Radiotherapy Response in Mice. Clin Oncol (R Coll Radiol) 2019; 31:272-282. [PMID: 30871751 DOI: 10.1016/j.clon.2019.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
Mouse models are essential tools in cancer research that have been used to understand the genetic basis of tumorigenesis, cancer progression and to test the efficacies of anticancer treatments including radiotherapy. They have played a critical role in our understanding of radiotherapy response in tumours and normal tissues and continue to evolve to better recapitulate the underlying biology of humans. In addition, recent developments in small animal irradiators have significantly improved in vivo irradiation techniques, allowing previously unimaginable experimental approaches to be explored in the laboratory. The combination of contemporary mouse models with small animal irradiators represents a major step forward for the radiobiology field in being able to much more accurately replicate clinical exposure scenarios. As radiobiology studies become ever more sophisticated in reflecting developments in the clinic, it is increasingly important to understand the basis and potential limitations of extrapolating data from mice to humans. This review provides an overview of mouse models and small animal radiotherapy platforms currently being used as advanced radiobiological research tools towards improving the translational power of preclinical studies.
Collapse
|
26
|
Azenha D, Lopes MC, Martins TC. Claspin: From replication stress and DNA damage responses to cancer therapy. DNA Repair (Amst) 2019; 115:203-246. [DOI: 10.1016/bs.apcsb.2018.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Mondal AM, Zhou H, Horikawa I, Suprynowicz FA, Li G, Dakic A, Rosenthal B, Ye L, Harris CC, Schlegel R, Liu X. Δ133p53α, a natural p53 isoform, contributes to conditional reprogramming and long-term proliferation of primary epithelial cells. Cell Death Dis 2018; 9:750. [PMID: 29970881 PMCID: PMC6030220 DOI: 10.1038/s41419-018-0767-7] [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: 02/12/2018] [Revised: 05/25/2018] [Accepted: 06/08/2018] [Indexed: 12/12/2022]
Abstract
We previously developed the technique of conditional reprogramming (CR), which allows primary epithelial cells from fresh or cryopreserved specimens to be propagated long-term in vitro, while maintaining their genetic stability and differentiation potential. This method requires a combination of irradiated fibroblast feeder cells and a Rho-associated kinase (ROCK) inhibitor. In the present study, we demonstrate increased levels of full-length p53 and its natural isoform, Δ133p53α, in conditionally reprogrammed epithelial cells from primary prostate, foreskin, ectocervical, and mammary tissues. Increased Δ133p53α expression is critical for CR since cell proliferation is rapidly inhibited following siRNA knockdown of endogenous Δ133p53α. Importantly, overexpression of Δ133p53α consistently delays the onset of cellular senescence of primary cells when cultured under non-CR conditions in normal keratinocyte growth medium (KGM). More significantly, the combination of Δ133p53α overexpression and ROCK inhibitor, without feeder cells, enables primary epithelial cells to be propagated long-term in vitro. We also show that Δ133p53α overexpression induces hTERT expression and telomerase activity and that siRNA knockdown of hTERT causes rapid inhibition of cell proliferation, indicating a critical role of hTERT for mediating the effects of Δ133p53α. Altogether, these data demonstrate a functional and regulatory link between p53 pathways and hTERT expression during the conditional reprogramming of primary epithelial cells.
Collapse
Affiliation(s)
- Abdul M Mondal
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA
| | - Hua Zhou
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA.,Guizhou Medical University, Guiyang, Guizhou, China
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Frank A Suprynowicz
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA
| | - Guangzhao Li
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA
| | - Aleksandra Dakic
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA
| | - Bernard Rosenthal
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA
| | - Lin Ye
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA.,Shenzhen Eye Hospital, Shenzhen, Guangdong, China
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard Schlegel
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA.
| | - Xuefeng Liu
- Center for Cell Reprograming, Department of Pathology, Georgetown University Medical Center, Georgrtown, WA, 20057, USA. .,Second Xianya Hospital (Adjunct Position), Zhongnan University, Changsha, Huna, China. .,Affiliated Cancer Hospital & Institute (Adjunct Position), Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
28
|
Zhu C, Martin HL, Crouch BT, Martinez AF, Li M, Palmer GM, Dewhirst MW, Ramanujam N. Near-simultaneous quantification of glucose uptake, mitochondrial membrane potential, and vascular parameters in murine flank tumors using quantitative diffuse reflectance and fluorescence spectroscopy. BIOMEDICAL OPTICS EXPRESS 2018; 9:3399-3412. [PMID: 29984105 PMCID: PMC6033552 DOI: 10.1364/boe.9.003399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/14/2018] [Accepted: 06/18/2018] [Indexed: 05/24/2023]
Abstract
The shifting metabolic landscape of aggressive tumors, with fluctuating oxygenation conditions and temporal changes in glycolysis and mitochondrial metabolism, is a critical phenomenon to study in order to understand negative treatment outcomes. Recently, we have demonstrated near-simultaneous optical imaging of mitochondrial membrane potential (MMP) and glucose uptake in non-tumor window chambers, using the fluorescent probes tetramethylrhodamine ethyl ester (TMRE) and 2-N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG). Here, we demonstrate a complementary technique to perform near-simultaneous in vivo optical spectroscopy of tissue vascular parameters, glucose uptake, and MMP in a solid tumor model that is most often used for therapeutic studies. Our study demonstrates the potential of optical spectroscopy as an effective tool to quantify the vascular and metabolic characteristics of a tumor, which is an important step towards understanding the mechanisms underlying cancer progression, metastasis, and resistance to therapies.
Collapse
Affiliation(s)
- Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Hannah L. Martin
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Amy F. Martinez
- Currently with Office of Research, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Martin Li
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Gregory M. Palmer
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University, Durham, NC 27710, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
29
|
Modeling radiation injury-induced cell death and countermeasure drug responses in a human Gut-on-a-Chip. Cell Death Dis 2018; 9:223. [PMID: 29445080 PMCID: PMC5833800 DOI: 10.1038/s41419-018-0304-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/13/2017] [Accepted: 01/12/2018] [Indexed: 12/24/2022]
Abstract
Studies on human intestinal injury induced by acute exposure to γ-radiation commonly rely on use of animal models because culture systems do not faithfully mimic human intestinal physiology. Here we used a human Gut-on-a-Chip (Gut Chip) microfluidic device lined by human intestinal epithelial cells and vascular endothelial cells to model radiation injury and assess the efficacy of radiation countermeasure drugs in vitro. Exposure of the Gut Chip to γ-radiation resulted in increased generation of reactive oxygen species, cytotoxicity, apoptosis, and DNA fragmentation, as well as villus blunting, disruption of tight junctions, and compromise of intestinal barrier integrity. In contrast, pre-treatment with a potential prophylactic radiation countermeasure drug, dimethyloxaloylglycine (DMOG), significantly suppressed all of these injury responses. Thus, the human Gut Chip may serve as an in vitro platform for studying radiation-induced cell death and associate gastrointestinal acute syndrome, in addition to screening of novel radio-protective medical countermeasure drugs.
Collapse
|
30
|
Perego P, Hempel G, Linder S, Bradshaw TD, Larsen AK, Peters GJ, Phillips RM. Cellular pharmacology studies of anticancer agents: recommendations from the EORTC-PAMM group. Cancer Chemother Pharmacol 2017; 81:427-441. [PMID: 29285635 DOI: 10.1007/s00280-017-3502-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/17/2017] [Indexed: 02/07/2023]
|
31
|
Citrin DE. Short-Term Screening Assays for the Identification of Therapeutics for Cancer. Cancer Res 2017; 76:3443-5. [PMID: 27306870 DOI: 10.1158/0008-5472.can-16-1299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, NIH, Bethesda, Maryland.
| |
Collapse
|
32
|
Cheah R, Srivastava R, Stafford ND, Beavis AW, Green V, Greenman J. Measuring the response of human head and neck squamous cell carcinoma to irradiation in a microfluidic model allowing customized therapy. Int J Oncol 2017; 51:1227-1238. [PMID: 28902347 DOI: 10.3892/ijo.2017.4118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/31/2017] [Indexed: 11/06/2022] Open
Abstract
Radiotherapy is the standard treatment for head and neck squamous cell carcinoma (HNSCC), however, radioresistance remains a major clinical problem despite significant improvements in treatment protocols. Therapeutic outcome could potentially be improved if a patient's tumour response to irradiation could be predicted ex vivo before clinical application. The present study employed a bespoke microfluidic device to maintain HNSCC tissue whilst subjecting it to external beam irradiation and measured the responses using a panel of cell death and proliferation markers. HNSCC biopsies from five newly-presenting patients [2 lymph node (LN); 3 primary tumour (PT)] were divided into parallel microfluidic devices and replicates of each tumour were subjected to single-dose irradiation (0, 5, 10, 15 and 20 Gy). Lactate dehydrogenase (LDH) release was measured and tissue sections were stained for cytokeratin (CK), cleaved-CK18 (cCK18), phosphorylated-H2AX (γH2AX) and Ki‑67 by immunohistochemistry. In addition, fragmented DNA was detected using terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL). Compared with non‑irradiated controls, higher irradiation doses resulted in elevated CK18-labelling index in two lymph nodes [15 Gy; 34.8% on LN1 and 31.7% on LN2 (p=0.006)] and a single laryngeal primary tumour (20 Gy; 31.5%; p=0.014). Significantly higher levels of DNA fragmentation were also detected in both lymph node samples and one primary tumour but at varying doses of irradiation, i.e., LN1 (20 Gy; 27.6%; p=0.047), LN2 (15 Gy; 15.3%; p=0.038) and PT3 (10 Gy; 35.2%; p=0.01). The γH2AX expression was raised but not significantly in the majority of samples. The percentage of Ki‑67 positive nuclei reduced dose-dependently following irradiation. In contrast no significant difference in LDH release was observed between irradiated groups and controls. There is clear inter- and intra-patient variability in response to irradiation when measuring a variety of parameters, which offers the potential for the approach to provide clinically valuable information.
Collapse
Affiliation(s)
- Ramsah Cheah
- Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | | | | | - Andrew W Beavis
- Radiation Physics, Hull and East Yorkshire Hospitals NHS Trust, Faculty of Science and Engineering, University of Hull, Hull, HU6 7RX, UK
| | - Victoria Green
- School of Life Sciences, University of Hull, Hull, HU6 7RX, UK
| | - John Greenman
- School of Life Sciences, University of Hull, Hull, HU6 7RX, UK
| |
Collapse
|
33
|
Exposing primary rat retina cell cultures to γ-rays: An in vitro model for evaluating radiation responses. Exp Eye Res 2017; 166:21-28. [PMID: 28958589 DOI: 10.1016/j.exer.2017.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 11/22/2022]
Abstract
Retinal tissue can receive incidental γ-rays exposure during radiotherapy either of tumors of the eye and optic nerve or of head-and-neck tumors, and during medical diagnostic procedures. Healthy retina is therefore at risk of suffering radiation-related side effects and the knowledge of pathophysiological response of retinal cells to ionizing radiations could be useful to design possible strategies of prevention and management of radiotoxicity. In this study, we have exploited an in vitro model (primary rat retinal cell culture) to study an array of biological effects induced on retinal neurons by γ-rays. Most of the different cell types present in retinal tissue - either of the neuronal or glial lineages - are preserved in primary rat retinal cultures. Similar to the retina in situ, neuronal cells undergo in vitro a maturational development shown by the formation of polarized neuritic trees and operating synapses. Since 2 Gy is the incidental dose received by the healthy retina per fraction when the standard treatment is delivered to the brain, retina cell cultures have been exposed to 1 or 2 Gy of γ-rays at different level of neuronal differentiation in vitro: days in vitro (DIV)2 or DIV8. At DIV9, retinal cultures were analyzed in terms of viability, apoptosis and characterized by immunocytochemistry to identify alterations in neuronal differentiation. After irradiation at DIV2, MTT assay revealed an evident loss of cell viability and βIII-tubulin immunostaining highlighted a marked neuritic damage, indicating that survived neurons showed an impaired differentiation. Differentiated cultures (DIV8) appeared to be more resistant with respect to undifferentiated, DIV2 cultures, both in terms of cell viability and differentiation. Apoptosis evaluated with TUNEL assay showed that irradiation at both DIV2 and DIV8 induced a significant increase in the apoptotic rate. To further investigate the effects of γ-rays on retinal neurons, we evaluated the expression of synaptic proteins, such as SNAP25 and synaptophysin. WB and immunofluorescence analysis showed an altered expression of these proteins in particular when cultures were irradiated at DIV2. To evaluate the effect of γ-rays on photoreceptors, we studied the expression of rhodopsin in WB analysis and immunofluorescence. Our results confirm data from the literature that differentiated photoreceptors appear to be more resistant to irradiation respect to other retinal cell types present in cultures. The results obtained suggest that γ-rays exposure of primary retinal cultures may contribute to shed further light on the mechanisms involved in γ-radiation-induced neurodegeneration.
Collapse
|
34
|
Schlesinger D, Lee M, Ter Haar G, Sela B, Eames M, Snell J, Kassell N, Sheehan J, Larner JM, Aubry JF. Equivalence of cell survival data for radiation dose and thermal dose in ablative treatments: analysis applied to essential tremor thalamotomy by focused ultrasound and gamma knife. Int J Hyperthermia 2017; 33:401-410. [PMID: 28044461 PMCID: PMC6203314 DOI: 10.1080/02656736.2016.1278281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Thermal dose and absorbed radiation dose have historically been difficult to compare because different biological mechanisms are at work. Thermal dose denatures proteins and the radiation dose causes DNA damage in order to achieve ablation. The purpose of this paper is to use the proportion of cell survival as a potential common unit by which to measure the biological effect of each procedure. Survival curves for both thermal and radiation doses have been extracted from previously published data for three different cell types. Fits of these curves were used to convert both thermal and radiation dose into the same quantified biological effect: fraction of surviving cells. They have also been used to generate and compare survival profiles from the only indication for which clinical data are available for both focused ultrasound (FUS) thermal ablation and radiation ablation: essential tremor thalamotomy. All cell types could be fitted with coefficients of determination greater than 0.992. As an illustration, survival profiles of clinical thalamotomies performed by radiosurgery and FUS are plotted on a same graph for the same metric: fraction of surviving cells. FUS and Gamma Knife have the potential to be used in combination to deliver a more effective treatment (for example, FUS may be used to debulk the main tumour mass, and radiation to treat the surrounding tumour bed). In this case, a model which compares thermal and radiation treatments is valuable in order to adjust the dose between the two.
Collapse
Affiliation(s)
- D Schlesinger
- a Department of Radiation Oncology , University of Virginia , Charlottesville , VA , USA
- c Department of Neurosurgery , University of Virginia , Charlottesville , VA , USA
| | - M Lee
- b Focused Ultrasound Foundation , Charlottesville , VA , USA
| | - G Ter Haar
- d Division of Radiotherapy and Imaging , The Institute of Cancer Research:Royal Marsden Hospital , London , UK
| | - B Sela
- b Focused Ultrasound Foundation , Charlottesville , VA , USA
| | - M Eames
- b Focused Ultrasound Foundation , Charlottesville , VA , USA
| | - J Snell
- b Focused Ultrasound Foundation , Charlottesville , VA , USA
- c Department of Neurosurgery , University of Virginia , Charlottesville , VA , USA
| | - N Kassell
- b Focused Ultrasound Foundation , Charlottesville , VA , USA
- c Department of Neurosurgery , University of Virginia , Charlottesville , VA , USA
| | - J Sheehan
- a Department of Radiation Oncology , University of Virginia , Charlottesville , VA , USA
- c Department of Neurosurgery , University of Virginia , Charlottesville , VA , USA
| | - J M Larner
- a Department of Radiation Oncology , University of Virginia , Charlottesville , VA , USA
| | - J-F Aubry
- a Department of Radiation Oncology , University of Virginia , Charlottesville , VA , USA
- e ESPCI Paris, PSL Research University, CNRS, INSERM, Institut Langevin , Paris , France
| |
Collapse
|
35
|
Liu X, Krawczyk E, Suprynowicz FA, Palechor-Ceron N, Yuan H, Dakic A, Simic V, Zheng YL, Sripadhan P, Chen C, Lu J, Hou TW, Choudhury S, Kallakury B, Tang DG, Darling T, Thangapazham R, Timofeeva O, Dritschilo A, Randell SH, Albanese C, Agarwal S, Schlegel R. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat Protoc 2017; 12:439-451. [PMID: 28125105 PMCID: PMC6195120 DOI: 10.1038/nprot.2016.174] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Historically, it has been difficult to propagate cells in vitro that are derived directly from human tumors or healthy tissue. However, in vitro preclinical models are essential tools for both the study of basic cancer biology and the promotion of translational research, including drug discovery and drug target identification. This protocol describes conditional reprogramming (CR), which involves coculture of irradiated mouse fibroblast feeder cells with normal and tumor human epithelial cells in the presence of a Rho kinase inhibitor (Y-27632). CR cells can be used for various applications, including regenerative medicine, drug sensitivity testing, gene expression profiling and xenograft studies. The method requires a pathologist to differentiate healthy tissue from tumor tissue, and basic tissue culture skills. The protocol can be used with cells derived from both fresh and cryopreserved tissue samples. As approximately 1 million cells can be generated in 7 d, the technique is directly applicable to diagnostic and predictive medicine. Moreover, the epithelial cells can be propagated indefinitely in vitro, yet retain the capacity to become fully differentiated when placed into conditions that mimic their natural environment.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Correspondence should be addressed to X.L. () or R.S. ()
| | - Ewa Krawczyk
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Correspondence should be addressed to X.L. () or R.S. ()
| | - Frank A Suprynowicz
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Nancy Palechor-Ceron
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Hang Yuan
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Aleksandra Dakic
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Vera Simic
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Yun-Ling Zheng
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Praathibha Sripadhan
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Chen Chen
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Jie Lu
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Tung-Wei Hou
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Sujata Choudhury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Bhaskar Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Dean G Tang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, Texas, USA
| | - Thomas Darling
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Rajesh Thangapazham
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Olga Timofeeva
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Anatoly Dritschilo
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Christopher Albanese
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Seema Agarwal
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
- Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
36
|
McMillan KS, Boyd M, Zagnoni M. Transitioning from multi-phase to single-phase microfluidics for long-term culture and treatment of multicellular spheroids. LAB ON A CHIP 2016; 16:3548-3557. [PMID: 27477673 DOI: 10.1039/c6lc00884d] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
When compared to methodologies based on low adhesion or hanging drop plates, droplet microfluidics offers several advantages for the formation and culture of multicellular spheroids, such as the potential for higher throughput screening and the use of reduced cell numbers, whilst providing increased stability for plate handling. However, a drawback of the technology is its characteristic compartmentalisation which limits the nutrients available to cells within an emulsion and poses challenges to the exchange of the encapsulated solution, often resulting in short-term cell culture and/or viability issues. The aim of this study was to develop a multi-purpose microfluidic platform that combines the high-throughput characteristics of multi-phase flows with that of ease of perfusion typical of single-phase microfluidics. We developed a versatile system to upscale the formation and long-term culture of multicellular spheroids for testing anticancer treatments, creating an array of fluidically addressable, compact spheroids that could be cultured in either medium or within a gel scaffold. The work provides proof-of-concept results for using this system to test both chemo- and radio-therapeutic protocols using in vitro 3D cancer models.
Collapse
Affiliation(s)
- Kay S McMillan
- Centre for Microsystems and Photonics, Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, UK.
| | | | | |
Collapse
|
37
|
Coleman CN, Higgins GS, Brown JM, Baumann M, Kirsch DG, Willers H, Prasanna PGS, Dewhirst MW, Bernhard EJ, Ahmed MM. Improving the Predictive Value of Preclinical Studies in Support of Radiotherapy Clinical Trials. Clin Cancer Res 2016; 22:3138-47. [PMID: 27154913 PMCID: PMC4930691 DOI: 10.1158/1078-0432.ccr-16-0069] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 01/16/2023]
Abstract
There is an urgent need to improve reproducibility and translatability of preclinical data to fully exploit opportunities for molecular therapeutics involving radiation and radiochemotherapy. For in vitro research, the clonogenic assay remains the current state-of-the-art of preclinical assays, whereas newer moderate and high-throughput assays offer the potential for rapid initial screening. Studies of radiation response modification by molecularly targeted agents can be improved using more physiologic 3D culture models. Elucidating effects on the cancer stem cells (CSC, and CSC-like) and developing biomarkers for defining targets and measuring responses are also important. In vivo studies are necessary to confirm in vitro findings, further define mechanism of action, and address immunomodulation and treatment-induced modification of the microenvironment. Newer in vivo models include genetically engineered and patient-derived xenograft mouse models and spontaneously occurring cancers in domesticated animals. Selection of appropriate endpoints is important for in vivo studies; for example, regrowth delay measures bulk tumor killing, whereas local tumor control assesses effects on CSCs. The reliability of individual assays requires standardization of procedures and cross-laboratory validation. Radiation modifiers must be tested as part of clinical standard of care, which includes radiochemotherapy for most tumors. Radiation models are compatible with but also differ from those used for drug screening. Furthermore, the mechanism of a drug as a chemotherapeutic agent may be different from its interaction with radiation and/or radiochemotherapy. This provides an opportunity to expand the use of molecular-targeted agents. Clin Cancer Res; 22(13); 3138-47. ©2016 AACR.
Collapse
Affiliation(s)
- C Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland.
| | - Geoff S Higgins
- Cancer Research UK/Medical Research Council, Oxford Institute for Radiation Oncology, University of Oxford, United Kingdom
| | - J Martin Brown
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Michael Baumann
- OncoRay National Center for Radiation Research, Technische Universität Dresden/Helmholtz-Zenrtum Dresden-Rossendorf, Dresden, Germany and German Cancer Consortium, Dresden/German Cancer Research Center (DKFZ)
| | - David G Kirsch
- Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pataje G S Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| | - Mark W Dewhirst
- Departments of Radiation Oncology, Pathology and Biomedical Engineering, Duke University, Durham, North Carolina
| | - Eric J Bernhard
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| | - Mansoor M Ahmed
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| |
Collapse
|
38
|
Stone HB, Bernhard EJ, Coleman CN, Deye J, Capala J, Mitchell JB, Brown JM. Preclinical Data on Efficacy of 10 Drug-Radiation Combinations: Evaluations, Concerns, and Recommendations. Transl Oncol 2016; 9:46-56. [PMID: 26947881 PMCID: PMC4800059 DOI: 10.1016/j.tranon.2016.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Clinical testing of new therapeutic interventions requires comprehensive, high-quality preclinical data. Concerns regarding quality of preclinical data have been raised in recent reports. This report examines the data on the interaction of 10 drugs with radiation and provides recommendations for improving the quality, reproducibility, and utility of future studies. The drugs were AZD6244, bortezomib, 17-DMAG, erlotinib, gefitinib, lapatinib, oxaliplatin/Lipoxal, sunitinib (Pfizer, Corporate headquarters, New York, NY), thalidomide, and vorinostat. METHODS In vitro and in vivo data were tabulated from 125 published papers, including methods, radiation and drug doses, schedules of administration, assays, measures of interaction, presentation and interpretation of data, dosimetry, and conclusions. RESULTS In many instances, the studies contained inadequate or unclear information that would hamper efforts to replicate or intercompare the studies, and that weakened the evidence for designing and conducting clinical trials. The published reports on these drugs showed mixed results on enhancement of radiation response, except for sunitinib, which was ineffective. CONCLUSIONS There is a need for improved experimental design, execution, and reporting of preclinical testing of agents that are candidates for clinical use in combination with radiation. A checklist is provided for authors and reviewers to ensure that preclinical studies of drug-radiation combinations meet standards of design, execution, and interpretation, and report necessary information to ensure high quality and reproducibility of studies. Improved design, execution, common measures of enhancement, and consistent interpretation of preclinical studies of drug-radiation interactions will provide rational guidance for prioritizing drugs for clinical radiotherapy trials and for the design of such trials.
Collapse
Affiliation(s)
- Helen B Stone
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - Eric J Bernhard
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727.
| | - C Norman Coleman
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - James Deye
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - Jacek Capala
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - James B Mitchell
- Radiation Biology Branch, National Cancer Institute, MSC 1002, 10 Center Dr, Bethesda, MD, 20892
| | - J Martin Brown
- Stanford University, Radiation and Cancer Biology, CCSR-S Rm 1255, 269 Campus Dr, Stanford, CA, 94305
| |
Collapse
|
39
|
McMillan KS, McCluskey AG, Sorensen A, Boyd M, Zagnoni M. Emulsion technologies for multicellular tumour spheroid radiation assays. Analyst 2016; 141:100-10. [DOI: 10.1039/c5an01382h] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Emulsion technology is used to assess the cytotoxic effect of radiation on spheroids by mimicking tumour growth and tumour quiescence.
Collapse
Affiliation(s)
- Kay S. McMillan
- Centre for Microsystems and Photonics
- Electronic and Electrical Engineering
- University of Strathclyde
- Glasgow
- UK
| | - Anthony G. McCluskey
- Strathclyde Institute of Pharmacy and Biomedical Sciences
- University of Strathclyde
- Glasgow
- UK
| | - Annette Sorensen
- Strathclyde Institute of Pharmacy and Biomedical Sciences
- University of Strathclyde
- Glasgow
- UK
| | - Marie Boyd
- Strathclyde Institute of Pharmacy and Biomedical Sciences
- University of Strathclyde
- Glasgow
- UK
| | - Michele Zagnoni
- Centre for Microsystems and Photonics
- Electronic and Electrical Engineering
- University of Strathclyde
- Glasgow
- UK
| |
Collapse
|
40
|
Cox MC, Reese LM, Bickford LR, Verbridge SS. Toward the Broad Adoption of 3D Tumor Models in the Cancer Drug Pipeline. ACS Biomater Sci Eng 2015; 1:877-894. [PMID: 33429520 DOI: 10.1021/acsbiomaterials.5b00172] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite a cost of approximately $1 billion to develop a new cancer drug, about 90% of drugs that enter clinical trials fail. A tremendous opportunity exists to streamline the drug selection and testing process, and innovative approaches promise to reduce the burdensome cost of health care for those suffering from cancer. There is great potential for 3D models of human tumors to complement more traditional testing methods; however, the shift from 2D to 3D assays at early stages of the drug discovery and development process is far from widely accepted. 3D platforms range from simple tumor spheroids to more complex microfluidic hydrogels that better mimic the tumor microenvironment. While several companies have developed and patented advanced high-throughput 3D platforms for drug screening, their cost and complexity have limited their adoption as an industry standard. In this review, we will highlight the various tumor platforms that have been developed, emphasizing the approaches that have successfully led to commercial products. We will then consider potential directions toward more relevant tumor models, advantages of the adoption of such platforms within the drug development and screening process, and new opportunities in personalized medicine that such platforms will uniquely enable.
Collapse
Affiliation(s)
- Megan C Cox
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| | - Laura M Reese
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| | - Lissett R Bickford
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| | - Scott S Verbridge
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, Virginia 24061, United States
| |
Collapse
|
41
|
Yu T, Luo L, Wang L. Ultrasound as a cancer chemotherapy sensitizer: the gap between laboratory and bedside. Expert Opin Drug Deliv 2015; 13:37-47. [DOI: 10.1517/17425247.2015.1083008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
42
|
Slabbert J, Martinez J, De Coster BM, Gueulette J. Increased Proton Relative Biological Effectiveness at the Very End of a Spread-Out Bragg Peak for Jejunum Irradiated Ex Vivo. Int J Part Ther 2015. [DOI: 10.14338/ijpt-14-00027.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
43
|
Ree AH, Redalen KR. Personalized radiotherapy: concepts, biomarkers and trial design. Br J Radiol 2015; 88:20150009. [PMID: 25989697 DOI: 10.1259/bjr.20150009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the past decade, and pointing onwards to the immediate future, clinical radiotherapy has undergone considerable developments, essentially including technological advances to sculpt radiation delivery, the demonstration of the benefit of adding concomitant cytotoxic agents to radiotherapy for a range of tumour types and, intriguingly, the increasing integration of targeted therapeutics for biological optimization of radiation effects. Recent molecular and imaging insights into radiobiology will provide a unique opportunity for rational patient treatment, enabling the parallel design of next-generation trials that formally examine the therapeutic outcome of adding targeted drugs to radiation, together with the critically important assessment of radiation volume and dose-limiting treatment toxicities. In considering the use of systemic agents with presumed radiosensitizing activity, this may also include the identification of molecular, metabolic and imaging markers of treatment response and tolerability, and will need particular attention on patient eligibility. In addition to providing an overview of clinical biomarker studies relevant for personalized radiotherapy, this communication will highlight principles in addressing clinical evaluation of combined-modality-targeted therapeutics and radiation. The increasing number of translational studies that bridge large-scale omics sciences with quality-assured phenomics end points-given the imperative development of open-source data repositories to allow investigators the access to the complex data sets-will enable radiation oncology to continue to position itself with the highest level of evidence within existing clinical practice.
Collapse
Affiliation(s)
- A H Ree
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,2 Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K R Redalen
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
44
|
Willey CD, Gilbert AN, Anderson JC, Gillespie GY. Patient-Derived Xenografts as a Model System for Radiation Research. Semin Radiat Oncol 2015; 25:273-80. [PMID: 26384275 DOI: 10.1016/j.semradonc.2015.05.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cancer literature is filled with promising preclinical studies demonstrating impressive efficacy for new therapeutics, yet translation of these approaches into clinical successes has been rare, indicating that current methods used to predict efficacy are suboptimal. The most likely reason for the limitation of these studies is the disconnect between preclinical models and cancers treated in the clinic. Specifically, most preclinical models are poor representations of human disease. Immortalized cancer cell lines that dominate the cancer literature may be, in a sense, "paper tigers" that have been selected by decades of culture to be artificially driven by highly targetable proteins. Thus, although effective in treating these cell lines either in vitro or as artificial tumors transplanted from culture into experimental animals as xenografts, the identified therapies would likely underperform in a clinical setting. This inherent limitation applies not only to drug testing but also to experiments with radiation therapy. Indeed, traditional radiobiology methods rely on monolayer culture systems, with emphasis on colony formation and DNA damage assessment that may have limited clinical translation. As such, there has been keen interest in developing tumor explant systems in which patient tumors are directly transplanted into and solely maintained in vivo, using immunocompromised mice. These so-called patient-derived xenografts (PDXs) represent a robust model system that has been garnering support in academia and industry as a superior preclinical approach to drug testing. Likewise, PDX models have the potential to improve radiation research. In this review, we describe how PDX models are currently being used for both drug and radiation testing and how they can be incorporated into a translational research program.
Collapse
Affiliation(s)
| | - Ashley N Gilbert
- Department of Radiation Oncology, The University of Alabama at Birmingham
| | - Joshua C Anderson
- Department of Radiation Oncology, The University of Alabama at Birmingham
| | | |
Collapse
|
45
|
Liu Q, Wang M, Kern AM, Khaled S, Han J, Yeap BY, Hong TS, Settleman J, Benes CH, Held KD, Efstathiou JA, Willers H. Adapting a drug screening platform to discover associations of molecular targeted radiosensitizers with genomic biomarkers. Mol Cancer Res 2015; 13:713-20. [PMID: 25667133 DOI: 10.1158/1541-7786.mcr-14-0570] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/19/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED Large collections of annotated cancer cell lines are powerful tools for precisely matching targeted drugs with genomic alterations that can be tested as biomarkers in the clinic. Whether these screening platforms, which utilize short-term cell survival to assess drug responses, can be applied to precision radiation medicine is not established. To this end, 32 cancer cell lines were screened using 18 targeted therapeutic agents with known or putative radiosensitizing properties (227 combinations). The cell number remaining after drug exposure with or without radiation was assessed by nonclonogenic assays. We derived short-term radiosensitization factors (SRF2Gy) and calculated clonogenic survival assay-based dose enhancement factors (DEFSF0.1). Radiosensitization was characterized by SRF2Gy values of mostly ∼1.05 to 1.2 and significantly correlated with drug-induced changes in apoptosis and senescence frequencies. SRF2Gy was significantly correlated with DEFSF0.1, with a respective sensitivity and specificity of 91.7% and 81.5% for a 3-day endpoint, and 82.8% and 84.2% for a robotic 5-day assay. KRAS mutations (codons 12/13) were found to be a biomarker of radiosensitization by midostaurin in lung cancer, which was pronounced under conditions that enriched for stem cell-like cells. In conclusion, although short-term proliferation/survival assays cannot replace the gold-standard clonogenic survival assay for measuring cellular radiosensitivity, they capture with high accuracy the relative change in radiosensitivity that is caused by a radiosensitzing targeted agent. IMPLICATIONS This study supports a paradigm shift regarding the utility of short-term assays for precision radiation medicine, which should facilitate the identification of genomic biomarkers to guide the testing of novel drug/radiation combinations.
Collapse
Affiliation(s)
- Qi Liu
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Meng Wang
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ashley M Kern
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Saman Khaled
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jing Han
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Jinan Municipal Center for Disease Control and Prevention, Shandong, China
| | - Beow Y Yeap
- Biostatistics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeff Settleman
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Cyril H Benes
- Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Kathryn D Held
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jason A Efstathiou
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Henning Willers
- Laboratory of Cellular and Molecular Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts. Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
46
|
Westwood JA, Potdevin Hunnam TCU, Pegram HJ, Hicks RJ, Darcy PK, Kershaw MH. Routes of delivery for CpG and anti-CD137 for the treatment of orthotopic kidney tumors in mice. PLoS One 2014; 9:e95847. [PMID: 24788789 PMCID: PMC4008493 DOI: 10.1371/journal.pone.0095847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 04/01/2014] [Indexed: 12/24/2022] Open
Abstract
We have found previously that the tumor cell lines, Renca (a renal cancer) and MC38 (a colon tumor) which had been injected subcutaneously in mice, could be successfully treated with a combination therapy of an oligodeoxynucleotide (CpG1826) (injected intratumorally) and anti-CD137 antibody (injected intraperitoneally). Thus the combination treatment was expected to initiate a “danger” signal via TLR9 on immune cells, and the anti-CD137 was expected to further activate T cells. In the present study, we found that several other tumor types injected subcutaneously could also be successfully treated with this combination therapy. In addition, we wished to determine if the treatment could work as effectively in an orthotopic metastatic model, which is more physiologically relevant to cancer in humans. Renca was selected as we were familiar with injecting this orthotopically into the outer cortex of the kidney in mice, and it spontaneously metastasizes to lung and abdominal sites. We tested various routes of delivery of CpG combined with intraperitoneal delivery of anti-CD137. Orthotopic tumors were injected with CpG intratumorally, using ultrasound-guided delivery on multiple occasions, combined with anti-CD137 intraperitoneally. A reduction in primary tumor size was observed following intratumoral injection of CpG compared to other treatments. We found that there was a statistically significant increase in survival of mice with orthotopic Renca tumor following intratumoral injection of CpG. However, we determined that the most effective route of delivery of CpG was intravenous, which led to further significantly enhanced survival of mice when combined with anti-CD137 intraperitoneally, likely due to inhibition of metastatic disease. Our data supports future development of this combination therapy for cancer.
Collapse
Affiliation(s)
- Jennifer A. Westwood
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | | | - Hollie J. Pegram
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Rodney J. Hicks
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Phillip K. Darcy
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
| | - Michael H. Kershaw
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
- * E-mail:
| |
Collapse
|
47
|
Abstract
Chemoradiation is the standard therapy for the majority of inoperable, locally advanced cancers. Although there is a need to improve chemoradiation efficacy, normal-tissue toxicity limits our ability to give additional chemotherapy or higher doses of radiation. Thus, there is excitement about the addition of molecularly targeted agents, which tend to be less toxic than chemotherapy, to chemoradiation regimens. Unfortunately, initial empiric attempts have not been successful. This review will focus on the evidence that supports rational combinations of targeted agents with chemoradiation, with an emphasis on agents that target the DNA damage response and radiation-induced membrane signaling.
Collapse
Affiliation(s)
- Meredith A. Morgan
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI
| | - Leslie A. Parsels
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Jonathan Maybaum
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - Theodore S. Lawrence
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
48
|
Nanoparticles targeting mechanisms in cancer therapy: current limitations and emerging solutions. Ther Deliv 2013; 4:1197-209. [DOI: 10.4155/tde.13.75] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
It has been more than one century since Paul Ehrlich spoke about the idea of targeting specific molecules in the cell when he coined the ‘Magic Bullet‘ principle. In most occasions, we seek new pharmacodynamic models for therapy, but nanoparticles provide a chance to modify the already existing pharmacokinetics of drugs to meet needed pharmacodynamic models. In the scope of ‘nanoscale‘, every entity has different characters, and no general rules control pharmacokinetics of nanoparticulate drugs as new physical and physicochemical properties are added to equations. However, such remarkable drug models are still quite far from achieving their potential in clinical application. Among the major obstacles is that most available results in nanoparticles targeting rely upon in vitro and animal models that do not match the tumor environment characteristics in humans. This Review discusses the concept of targeting tumor cells with nanoparticles, the limitations that lead to its incomplete application in clinical practice along with some of the promising solutions to such limitations.
Collapse
|