1
|
Pediatric Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00075-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
2
|
Damaraju VL, Aminpour M, Kuzma M, Winter P, Preto J, Tuszynski J, McEwan ABJ, Sawyer MB. Tyrosine Kinase Inhibitors Reduce Glucose Uptake by Binding to an Exofacial Site on hGLUT-1: Influence on 18 F-FDG PET Uptake. Clin Transl Sci 2020; 14:847-858. [PMID: 33278334 PMCID: PMC8212708 DOI: 10.1111/cts.12943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
Positron emission tomography (PET) using 2‐deoxy‐2‐[18F]fluoro‐d‐glucose ([18F]FDG), a marker of energy metabolism and cell proliferation, is routinely used in the clinic to assess patient response to chemotherapy and to monitor tumor growth. Treatment with some tyrosine kinase inhibitors (TKIs) causes changes in blood glucose levels in both nondiabetic and diabetic patients. We evaluated the interaction of several classes of TKIs with human glucose transporter‐1 (hGLUT‐1) in FaDu and GIST‐1 cells by measuring [3H]2‐deoxy‐d‐glucose ([3H]2‐DG) and [3H]FDG uptake. Uptake of both was inhibited to varying extents by the TKIs, and representative TKIs from each class showed competitive inhibition of [3H]2‐DG uptake. In GIST‐1 cells, [3H]FDG uptake inhibition by temsirolimus and nilotinib was irreversible, whereas inhibition by imatinib, gefitinib, and pazopanib was reversible. Molecular modeling studies showed that TKIs form multiple hydrogen bonds with polar residues of the sugar binding site (i.e., Q161, Q282, Q283, N288, N317, and W388), and van der Waals interactions with the H‐pocket site. Our results showed interaction of TKIs with amino acid residues at the glucose binding site to inhibit glucose uptake by hGLUT‐1. We hypothesize that inhibition of hGLUT‐1 by TKIs could alter glucose levels in patients treated with TKIs, leading to hypoglycemia and fatigue, although further studies are required to evaluate roles of other SLC2 and SLC5 members. In addition, TKIs could affect tumor [18F]FDG uptake, increasingly used as a marker of tumor response. The hGLUT‐1 inhibition by TKIs may have implications for routine [18F]FDG‐PET monitoring of tumor response in patients.
Collapse
Affiliation(s)
- Vijaya L Damaraju
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Maral Aminpour
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Michelle Kuzma
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Philip Winter
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Jordane Preto
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada.,DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi, Torino, Italy
| | - Jack Tuszynski
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander B J McEwan
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Michael B Sawyer
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Theruvath AJ, Siedek F, Muehe AM, Garcia-Diaz J, Kirchner J, Martin O, Link MP, Spunt S, Pribnow A, Rosenberg J, Herrmann K, Gatidis S, Schäfer JF, Moseley M, Umutlu L, Daldrup-Link HE. Therapy Response Assessment of Pediatric Tumors with Whole-Body Diffusion-weighted MRI and FDG PET/MRI. Radiology 2020; 296:143-151. [PMID: 32368961 PMCID: PMC7325702 DOI: 10.1148/radiol.2020192508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 12/26/2022]
Abstract
Background Whole-body diffusion-weighted (DW) MRI can help detect cancer with high sensitivity. However, the assessment of therapy response often requires information about tumor metabolism, which is measured with fluorine 18 fluorodeoxyglucose (FDG) PET. Purpose To compare tumor therapy response with whole-body DW MRI and FDG PET/MRI in children and young adults. Materials and Methods In this prospective, nonrandomized multicenter study, 56 children and young adults (31 male and 25 female participants; mean age, 15 years ± 4 [standard deviation]; age range, 6-22 years) with lymphoma or sarcoma underwent 112 simultaneous whole-body DW MRI and FDG PET/MRI between June 2015 and December 2018 before and after induction chemotherapy (ClinicalTrials.gov identifier: NCT01542879). The authors measured minimum tumor apparent diffusion coefficients (ADCs) and maximum standardized uptake value (SUV) of up to six target lesions and assessed therapy response after induction chemotherapy according to the Lugano classification or PET Response Criteria in Solid Tumors. The authors evaluated agreements between whole-body DW MRI- and FDG PET/MRI-based response classifications with Krippendorff α statistics. Differences in minimum ADC and maximum SUV between responders and nonresponders and comparison of timing for discordant and concordant response assessments after induction chemotherapy were evaluated with the Wilcoxon test. Results Good agreement existed between treatment response assessments after induction chemotherapy with whole-body DW MRI and FDG PET/MRI (α = 0.88). Clinical response prediction according to maximum SUV (area under the receiver operating characteristic curve = 100%; 95% confidence interval [CI]: 99%, 100%) and minimum ADC (area under the receiver operating characteristic curve = 98%; 95% CI: 94%, 100%) were similar (P = .37). Sensitivity and specificity were 96% (54 of 56 participants; 95% CI: 86%, 99%) and 100% (56 of 56 participants; 95% CI: 54%, 100%), respectively, for DW MRI and 100% (56 of 56 participants; 95% CI: 93%, 100%) and 100% (56 of 56 participants; 95% CI: 54%, 100%) for FDG PET/MRI. In eight of 56 patients who underwent imaging after induction chemotherapy in the early posttreatment phase, chemotherapy-induced changes in tumor metabolism preceded changes in proton diffusion (P = .002). Conclusion Whole-body diffusion-weighted MRI showed significant agreement with fluorine 18 fluorodeoxyglucose PET/MRI for treatment response assessment in children and young adults. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ashok J. Theruvath
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Florian Siedek
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Anne M. Muehe
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Jordi Garcia-Diaz
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Julian Kirchner
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Ole Martin
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Michael P. Link
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Sheri Spunt
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Allison Pribnow
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Jarrett Rosenberg
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Ken Herrmann
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Sergios Gatidis
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Jürgen F. Schäfer
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Michael Moseley
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Lale Umutlu
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| | - Heike E. Daldrup-Link
- From the Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Rd, Stanford, CA 94304 (A.J.T., F.S., A.M.M., J.G.D., J.R., M.M., H.E.D.L.); Department of Diagnostic and Interventional Radiology, University Medical Center Mainz, Mainz, Germany (A.J.T.); Institute of Diagnostic and Interventional Radiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany (F.S.); Department of Diagnostic and Interventional Radiology, Medical Faculty, University Düsseldorf, Düsseldorf, Germany (J.K., O.M.); Department of Pediatrics, Pediatric Oncology, Lucile Packard Children’s Hospital, Stanford University, Stanford, Calif (M.P.L., S.S., A.P., H.E.D.L.); Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (K.H.); Department of Diagnostic and Interventional Radiology, University Hospital Tuebingen, Tuebingen, Germany (S.G., J.F.S.); Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany (L.U.)
| |
Collapse
|
4
|
Iommelli F, De Rosa V, Terlizzi C, Fonti R, Del Vecchio S. Preclinical Imaging in Targeted Cancer Therapies. Semin Nucl Med 2019; 49:369-381. [PMID: 31470932 DOI: 10.1053/j.semnuclmed.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Preclinical imaging with radiolabeled probes can provide noninvasive tools to test the efficacy of targeted agents in tumors harboring specific genetic alterations and to identify imaging parameters that can be used as pharmacodynamics markers in cancer patients. The present review will primarily focus on preclinical imaging studies that can accelerate the clinical approval of targeted agents and promote the development of imaging biomarkers for clinical applications. Since only subgroups of patients may benefit from treatment with targeted anticancer agents, the identification of a patient population expressing the target is of primary importance for the success of clinical trials. Preclinical imaging studies tested the ability of new radiolabeled compounds to recognize mutant, amplified, or overexpressed targets and some of these tracers were transferred to the clinical setting. More common tracers such as 18F-Fluorothymidine and 18F-Fluorodeoxyglucose were employed in animal models to test the inhibition of the target and downstream pathways through the evaluation of early changes of proliferation and glucose metabolism allowing the identification of sensitive and resistant tumors. Furthermore, since the majority of patients treated with targeted anticancer agents will invariably develop resistance, preclinical imaging studies were performed to test the efficacy of reversal agents to overcome resistance. These studies provided consistent evidence that imaging with radiolabeled probes can monitor the reversal of drug resistance by newly designed alternative compounds. Finally, despite many difficulties and challenges, preclinical imaging studies targeting the expression of immune checkpoints proved the principle that it is feasible to select patients for immunotherapy based on imaging findings. In conclusion, preclinical imaging can be considered as an integral part of the complex translational process that moves a newly developed targeted agent from laboratory to clinical application intervening in all clinically relevant steps including patient selection, early monitoring of drug effects and reversal of drug resistance.
Collapse
Affiliation(s)
- Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Cristina Terlizzi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Rosa Fonti
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
5
|
Ravoori MK, Singh SP, Lee J, Bankson JA, Kundra V. In Vivo Assessment of Ovarian Tumor Response to Tyrosine Kinase Inhibitor Pazopanib by Using Hyperpolarized 13C-Pyruvate MR Spectroscopy and 18F-FDG PET/CT Imaging in a Mouse Model. Radiology 2017; 285:830-838. [PMID: 28707963 DOI: 10.1148/radiol.2017161772] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To assess in a mouse model whether early or late components of glucose metabolism, exemplified by fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) and hyperpolarized carbon 13 (13C)-pyruvate magnetic resonance (MR) spectroscopy, can serve as indicators of response in ovarian cancer to multityrosine kinase inhibitor pazopanib. Materials and Methods In this Animal Care and Use Committee approved study, 17 days after the injection of 2 × 106 human ovarian SKOV3 tumors cells into 14 female nude mice, treatment with vehicle or pazopanib (2.5 mg per mouse peroral every other day) was initiated. Longitudinal T2-weighted MR imaging, dynamic MR spectroscopy of hyperpolarized pyruvate, and 18F-FDG PET/computed tomographic (CT) imaging were performed before treatment, 2 days after treatment, and 2 weeks after treatment. Results Pazopanib inhibited ovarian tumor growth compared with control (0.054 g ± 0.041 vs 0.223 g ± 0.112, respectively; six mice were treated with pazopanib and seven were control mice; P < .05). Significantly higher pyruvate-to-lactate conversion (lactate/pyruvate + lactate ratio) was found 2 days after treatment with pazopanib than before treatment (0.46 ± 0.07 vs 0.31 ± 0.14, respectively; P < .05; six tumors after treatment, seven tumors before treatment). This was not observed with the control group or with 18F-FDG PET/CT imaging. Conclusion The findings suggest that hyperpolarized 13C-pyruvate MR spectroscopy may serve as an early indicator of response to tyrosine kinase (angiogenesis) inhibitors such as pazopanib in ovarian cancer even when 18F-FDG PET/CT does not indicate a response. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Murali K Ravoori
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Sheela P Singh
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Jaehyuk Lee
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - James A Bankson
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Vikas Kundra
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| |
Collapse
|
6
|
Pinto F, Campanella NC, Abrahão-Machado LF, Scapulatempo-Neto C, de Oliveira AT, Brito MJ, Andrade RP, Guimarães DP, Reis RM. The embryonic Brachyury transcription factor is a novel biomarker of GIST aggressiveness and poor survival. Gastric Cancer 2016; 19:651-659. [PMID: 25995035 DOI: 10.1007/s10120-015-0505-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/29/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The T-box transcription factor Brachyury was recently reported to be upregulated and associated with prognosis in solid tumors. Here, we proposed to evaluate the potential use of Brachyury protein expression as a new prognostic biomarker in gastrointestinal stromal tumors (GIST). METHODS Brachyury protein expression was analyzed by immunohistochemistry in a cohort of 63 bona fide GIST patients. Brachyury expression profiles were correlated with patients' clinicopathological features and prognostic impact. Additionally, an in silico analysis was performed using the Oncomine database to assess Brachyury alterations at DNA and mRNA levels in GISTs. RESULTS We found that Brachyury was overexpressed in the majority (81.0 %) of primary GISTs. We observed Brachyury staining in the nucleus alone in 4.8 % of cases, 23.8 % depicted only cytoplasm staining, and 52.4 % of cases exhibited both nucleus and cytoplasm immunostaining. The presence of Brachyury was associated with aggressive GIST clinicopathological features. Particularly, Brachyury nuclear (with or without cytoplasm) staining was associated with the presence of metastasis, while cytoplasm sublocalization alone was correlated with poor patient survival. CONCLUSIONS Herein, we demonstrate that Brachyury is overexpressed in GISTs and is associated with worse outcome, constituting a novel prognostic biomarker and a putative target for GIST treatment.
Collapse
Affiliation(s)
- Filipe Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nathalia C Campanella
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784 400, Sao Paulo, Brazil
| | | | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784 400, Sao Paulo, Brazil
- Department of Pathology, Barretos Cancer Hospital, Barretos, Sao Paulo, Brazil
| | - Antonio T de Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784 400, Sao Paulo, Brazil
- Upper Digestive Surgery Department, Barretos Cancer Hospital, Barretos, Sao Paulo, Brazil
| | - Maria J Brito
- Department of Pathology, Hospital Garcia de Orta, Almada, Portugal
| | - Raquel P Andrade
- Regenerative Medicine Program, Department of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Denise P Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784 400, Sao Paulo, Brazil
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, Sao Paulo, Brazil
| | - Rui M Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784 400, Sao Paulo, Brazil.
| |
Collapse
|
7
|
Kinross KM, Montgomery KG, Mangiafico SP, Hare LM, Kleinschmidt M, Bywater MJ, Poulton IJ, Vrahnas C, Henneicke H, Malaterre J, Waring PM, Cullinane C, Sims NA, McArthur GA, Andrikopoulos S, Phillips WA. Ubiquitous expression of the Pik3caH1047R mutation promotes hypoglycemia, hypoinsulinemia, and organomegaly. FASEB J 2014; 29:1426-34. [PMID: 25550458 DOI: 10.1096/fj.14-262782] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/28/2014] [Indexed: 11/11/2022]
Abstract
Mutations in PIK3CA, the gene encoding the p110α catalytic subunit of PI3K, are among the most common mutations found in human cancer and have also recently been implicated in a range of overgrowth syndromes in humans. We have used a novel inducible "exon-switch" approach to knock in the constitutively active Pik3ca(H1047R) mutation into the endogenous Pik3ca gene of the mouse. Ubiquitous expression of the Pik3ca(H1047R) mutation throughout the body resulted in a dramatic increase in body weight within 3 weeks of induction (mutant 150 ± 5%; wild-type 117 ± 3%, mean ± sem), which was associated with increased organ size rather than adiposity. Severe metabolic effects, including a reduction in blood glucose levels to 59 ± 4% of baseline (11 days postinduction) and undetectable insulin levels, were also observed. Pik3ca(H1047R) mutant mice died earlier (median survival 46.5 d post-mutation induction) than wild-type control mice (100% survival > 250 days). Although deletion of Akt2 increased median survival by 44%, neither organ overgrowth, nor hypoglycemia were rescued, indicating that both the growth and metabolic functions of constitutive PI3K activity can be Akt2 independent. This mouse model demonstrates the critical role of PI3K in the regulation of both organ size and glucose metabolism at the whole animal level.
Collapse
Affiliation(s)
- Kathryn M Kinross
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Karen G Montgomery
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Salvatore P Mangiafico
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Lauren M Hare
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Margarete Kleinschmidt
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Megan J Bywater
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Ingrid J Poulton
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Christina Vrahnas
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Holger Henneicke
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Jordane Malaterre
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Paul M Waring
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Carleen Cullinane
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Natalie A Sims
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Grant A McArthur
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Sofianos Andrikopoulos
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Wayne A Phillips
- *Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Department of Medicine, Austin Health, Heidelberg, Victoria, Australia; Translational Research Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research and University of Melbourne Department of Medicine, St. Vincent's Hospital, Fitzroy, Victoria, Australia; Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; **Department of Pathology, University of Melbourne, Parkville, Victoria, Australia; Molecular Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; and University of Melbourne Department of Surgery, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| |
Collapse
|
8
|
Newbold A, Martin BP, Cullinane C, Bots M. Measuring apoptosis in mammals in vivo. Cold Spring Harb Protoc 2014; 2014:1125-1127. [PMID: 25368316 DOI: 10.1101/pdb.top070417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Apoptosis is a mode of cell death that is essential in multicellular organisms for the removal of superfluous, damaged, or potentially dangerous cells during development, infection, or normal tissue homeostasis. To prevent inflammation, cells undergoing apoptosis produce "find-me" signals that trigger the recruitment of phagocytes, which clear the apoptotic cells on recognition of "eat-me" signals. Despite the loss of billions of cells per day by apoptosis in the human body, the number of apoptotic cells found in healthy tissue is surprisingly low and reflects the efficiency of this process. However, in certain conditions (e.g., in cancer cells responding to chemotherapy), the number of apoptotic cells is too high to be efficiently cleared by phagocytes, and apoptotic cells can be observed. In these situations, the detection of apoptosis may be helpful in monitoring disease progression as well as in predicting the responses of tumors to anticancer therapies. Here we introduce various methods for monitoring apoptotic cells in vivo using a murine model of B-cell lymphoma and a solid tumor xenograft.
Collapse
Affiliation(s)
- Andrea Newbold
- Gene Regulation Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne 3002, Victoria, Australia
| | - Ben P Martin
- Gene Regulation Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne 3002, Victoria, Australia
| | - Carleen Cullinane
- Translational Research Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, East Melbourne 3002, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville 3010, Victoria, Australia
| | - Michael Bots
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine, Academic Medical Center, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Farwell MD, Pryma DA, Mankoff DA. PET/CT imaging in cancer: current applications and future directions. Cancer 2014; 120:3433-45. [PMID: 24947987 DOI: 10.1002/cncr.28860] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/05/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Positron emission tomography (PET) is a radiotracer imaging method that yields quantitative images of regional in vivo biology and biochemistry. PET, now used in conjunction with computed tomography (CT) in PET/CT devices, has had its greatest impact to date on cancer and is now an important part of oncologic clinical practice and translational cancer research. In this review of current applications and future directions for PET/CT in cancer, the authors first highlight the basic principles of PET followed by a discussion of the biochemistry and current clinical applications of the most commonly used PET imaging agent, (18) F-fluorodeoxyglucose (FDG). Then, emerging methods for PET imaging of other biologic processes relevant to cancer are reviewed, including cellular proliferation, tumor hypoxia, apoptosis, amino acid and cell membrane metabolism, and imaging of tumor receptors and other tumor-specific gene products. The focus of the review is on methods in current clinical practice as well as those that have been translated to patients and are currently in clinical trials.
Collapse
Affiliation(s)
- Michael D Farwell
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
10
|
Kanazu M, Maruyama K, Ando M, Asami K, Ishii M, Uehira K, Minomo S, Matsuda Y, Kawaguchi T, Atagi S, Ogawa Y, Kusunoki Y, Takada M, Kubo A. Early Pharmacodynamic Assessment Using 18F-Fluorodeoxyglucose Positron-Emission Tomography on Molecular Targeted Therapy and Cytotoxic Chemotherapy for Clinical Outcome Prediction. Clin Lung Cancer 2014; 15:182-7. [DOI: 10.1016/j.cllc.2014.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 11/15/2022]
|
11
|
Hensley H, Devarajan K, Johnson JR, Piwnica-Worms D, Godwin AK, von Mehren M, Rink L. Evaluating new therapies in gastrointestinal stromal tumor using in vivo molecular optical imaging. Cancer Biol Ther 2014; 15:911-8. [PMID: 24755645 DOI: 10.4161/cbt.28880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors in the US. The majority (~85%) of GISTs possess gain-of-function mutations in KIT or PDGFRA, causing constitutive activation of the kinase receptor. GIST management has been transformed by the identification of tumor driver mutations leading to unprecedented disease control of advanced GIST with the introduction of imatinib mesylate (IM). Despite IM's efficacy, most patients experience primary and/or secondary resistance within 2 y of treatment. Additional therapies and methods to optimize screening of novel approaches in preclinical studies are warranted. Clinically, treatment efficacy is typically assessed using Response Evaluation Criteria In Solid Tumors (RECIST) guidelines or Choi criteria. Both require a period of time on therapy before changes indicative of response can be observed. In addition, neither informs directly about cell death. We evaluated the use of molecular imaging technology in an animal model using near-infrared (NIR) imaging probes together with three-dimensional fluorescence molecular tomography (FMT) for assessing therapeutic response and ultimately optimizing our understanding of the biologic effects of these agents. We determined the potential of NIR probes (PSVue(TM) 794 and cell-penetrating KcapQ647) for detecting distinct markers of apoptosis and compare this to tumor size measured by MRI in response to IM treatment in GIST-T1 xenografts. Our studies revealed statistically significant increases in apoptosis due to IM treatment using both probes as early as 24 h post IM treatment which was confirmed by IHC. Molecular imaging will allow for faster and more effective screening of novel therapies in preclinical GIST models.
Collapse
Affiliation(s)
- Harvey Hensley
- Biological Imaging Facility; Fox Chase Cancer Center; Philadelphia, PA USA
| | - Karthik Devarajan
- Department of Statistics; Fox Chase Cancer Center; Philadelphia, PA USA
| | - James R Johnson
- Mallinckrodt Institute of Radiology; Washington University School of Medicine; St. Louis, MO USA
| | - David Piwnica-Worms
- Mallinckrodt Institute of Radiology; Washington University School of Medicine; St. Louis, MO USA; Department of Cancer Systems Imaging; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine; University of Kansas Medical Center; Kansas City, KS USA
| | - Margaret von Mehren
- Department of Medical Oncology; Fox Chase Cancer Center; Philadelphia, PA USA
| | - Lori Rink
- Department of Medical Oncology; Fox Chase Cancer Center; Philadelphia, PA USA
| |
Collapse
|
12
|
Tanaka M, Kataoka H, Yano S, Ohi H, Moriwaki K, Akashi H, Taguchi T, Hayashi N, Hamano S, Mori Y, Kubota E, Tanida S, Joh T. Antitumor Effects in Gastrointestinal Stromal Tumors Using Photodynamic Therapy with a Novel Glucose-Conjugated Chlorin. Mol Cancer Ther 2014; 13:767-75. [DOI: 10.1158/1535-7163.mct-13-0393] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Cullinane C, Solomon B, Hicks RJ. Imaging of molecular target modulation in oncology: challenges of early clinical trials. Clin Transl Imaging 2014. [DOI: 10.1007/s40336-013-0047-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
14
|
Preclinical evaluation of a novel c-Met inhibitor in a gastric cancer xenograft model using small animal PET. Mol Imaging Biol 2013; 15:203-11. [PMID: 22864665 DOI: 10.1007/s11307-012-0580-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Here, we describe the efficacy of the novel small molecule c-Met inhibitor BAY 853474 in reducing tumor growth in the Hs746T gastric cancer xenograft model and tested the suitability of 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG) versus 3'-deoxy-3'-18F-fluorothymidine ([(18)F]FLT) for response monitoring in a gastric cancer xenograft mouse model using small animal PET. PROCEDURES The c-Met inhibitor or vehicle control was administered orally at various doses in tumor-bearing mice. Glucose uptake and proliferation was measured using PET before, 48 and 96 h after the first treatment. The PET data were compared to data from tumor growth curves, autoradiography, Glut-1 and Ki-67 staining of tumor sections, and biochemical analysis of tissue probes, i.e., c-Met and ERK phosphorylation and cyclin D1 levels. RESULTS BAY 853474 significantly reduces tumor growth. [(18)F]FDG uptake in Hs746T tumors was significantly reduced in the groups receiving the drug, compared with the control group. The [(18)F]FLT uptake in the tumor tissue was completely absent 96 h after treatment. Autoradiographic, immunohistochemical, and biochemical analyses confirmed the PET findings. Treatment with the c-Met inhibitor did not affect body weight or glucose levels, and no adverse effects were observed in the animals. CONCLUSION These preclinical findings suggest that clinical PET imaging is a useful tool for early response monitoring in clinical studies.
Collapse
|
15
|
Kansara M, Leong HS, Lin DM, Popkiss S, Pang P, Garsed DW, Walkley CR, Cullinane C, Ellul J, Haynes NM, Hicks R, Kuijjer ML, Cleton-Jansen AM, Hinds PW, Smyth MJ, Thomas DM. Immune response to RB1-regulated senescence limits radiation-induced osteosarcoma formation. J Clin Invest 2013; 123:5351-60. [PMID: 24231354 DOI: 10.1172/jci70559] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 09/05/2013] [Indexed: 12/19/2022] Open
Abstract
Ionizing radiation (IR) and germline mutations in the retinoblastoma tumor suppressor gene (RB1) are the strongest risk factors for developing osteosarcoma. Recapitulating the human predisposition, we found that Rb1+/- mice exhibited accelerated development of IR-induced osteosarcoma, with a latency of 39 weeks. Initial exposure of osteoblasts to carcinogenic doses of IR in vitro and in vivo induced RB1-dependent senescence and the expression of a panel of proteins known as senescence-associated secretory phenotype (SASP), dominated by IL-6. RB1 expression closely correlated with that of the SASP cassette in human osteosarcomas, and low expression of both RB1 and the SASP genes was associated with poor prognosis. In vivo, IL-6 was required for IR-induced senescence, which elicited NKT cell infiltration and a host inflammatory response. Mice lacking IL-6 or NKT cells had accelerated development of IR-induced osteosarcomas. These data elucidate an important link between senescence, which is a cell-autonomous tumor suppressor response, and the activation of host-dependent cancer immunosurveillance. Our findings indicate that overcoming the immune response to senescence is a rate-limiting step in the formation of IR-induced osteosarcoma.
Collapse
|
16
|
Tegnebratt T, Lu L, Lee L, Meresse V, Tessier J, Ishii N, Harada N, Pisa P, Stone-Elander S. [18 F]FDG-PET imaging is an early non-invasive pharmacodynamic biomarker for a first-in-class dual MEK/Raf inhibitor, RO5126766 (CH5126766), in preclinical xenograft models. EJNMMI Res 2013; 3:67. [PMID: 24041012 PMCID: PMC3848680 DOI: 10.1186/2191-219x-3-67] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/08/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Positron emission tomography (PET) with [2-18 F]-2-fluoro-2-deoxy-D-glucose ([18 F]FDG-PET) was acquired at multiple time-points a) to monitor the early response to RO5126766 (CH5126766) in xenograft models b) to evaluate non-invasive small animal [18 F]FDG-PET imaging as a biomarker for MEK inhibitors for translation into dose-finding studies in cancer patients and c) to explore the underlying mechanism related to FDG uptake in tumors treated with RO5126766. METHODS [18 F]FDG uptake was studied in HCT116 (K-ras), COLO205 (B-raf) mutants and COLO320DM (wild type) xenografts from day 0 to 3 of RO5126766 treatment using a microPET Focus 120 and complemented with in vitro incubations, ex-vivo phosphor imaging and immunohistochemical (IHC) analyses. RESULTS In the HCT116 (K-ras) and COLO205 (B-raf) mutant xenografts, significant decreases in [18 F]FDG uptake were detected in vivo on day 1 with 0.3 mg/kg and ex vivo on day 3 with 0.1 mg/kg RO5126766. [18 F]FDG changes correlated with decreases in tumor cells proliferation (Ki-67) and with changes in expression levels of GLUT1. No effects were observed in drug resistant COLO320DM cells. The cellular fractionation and Western blotting analyses suggested that the change of [18 F]FDG uptake associated with RO5126766 is due to translocation of GLUT1 from membrane to cytosol, similar to the results reported in the literature with EGFR tyrosine kinase inhibitors, which also target the MAPK pathway. CONCLUSIONS RO5126766 inhibition resulted in a rapid time - and dose - dependent decline in [18 F]FDG uptake in both mutant xenografts. These results strongly resemble the clinical observations obtained with MEK/Raf inhibitors support the use of preclinical [18 F]FDG-PET as a translational tool for decision support in preclinical and early clinical development of MEK inhibitors.
Collapse
Affiliation(s)
- Tetyana Tegnebratt
- Neuro Fogrp Stone-Elander, Neuroradiology, K8, MicroPET and Clinical Neurosciences, H3:00, Karolinska University Hospital, Karolinska Institutet, Stockholm SE-17176, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Vettukattil R, Hetland TE, Flørenes VA, Kærn J, Davidson B, Bathen TF. Proton magnetic resonance metabolomic characterization of ovarian serous carcinoma effusions: chemotherapy-related effects and comparison with malignant mesothelioma and breast carcinoma. Hum Pathol 2013; 44:1859-66. [DOI: 10.1016/j.humpath.2013.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 02/09/2013] [Accepted: 02/11/2013] [Indexed: 10/26/2022]
|
18
|
Combined inhibition of PI3K-related DNA damage response kinases and mTORC1 induces apoptosis in MYC-driven B-cell lymphomas. Blood 2013; 121:2964-74. [PMID: 23403624 DOI: 10.1182/blood-2012-08-446096] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pharmacological strategies capable of directly targeting MYC are elusive. Previous studies have shown that MYC-driven lymphomagenesis is associated with mammalian target of rapamycin (mTOR) activation and a MYC-evoked DNA damage response (DDR) transduced by phosphatidylinositol-3-kinase (PI3K)-related kinases (DNA-PK, ATM, and ATR). Here we report that BEZ235, a multitargeted pan-PI3K/dual-mTOR inhibitor, potently killed primary Myc-driven B-cell lymphomas and human cell lines bearing IG-cMYC translocations. Using pharmacologic and genetic dissection of PI3K/mTOR signaling, dual DDR/mTORC1 inhibition was identified as a key mediator of apoptosis. Moreover, apoptosis was initiated at drug concentrations insufficient to antagonize PI3K/mTORC2-regulated AKT phosphorylation. p53-independent induction of the proapoptotic BH3-only protein BMF was identified as a mechanism by which dual DDR/mTORC1 inhibition caused lymphoma cell death. BEZ235 treatment induced apoptotic tumor regressions in vivo that correlated with suppression of mTORC1-regulated substrates and reduced H2AX phosphorylation and also with feedback phosphorylation of AKT. These mechanistic studies hold important implications for the use of multitargeted PI3K inhibitors in the treatment of hematologic malignancies. In particular, the newly elucidated role of PI3K-related DDR kinases in response to PI3K inhibitors offers a novel therapeutic opportunity for the treatment of hematologic malignancies with an MYC-driven DDR.
Collapse
|
19
|
Alberghina L, Gaglio D, Gelfi C, Moresco RM, Mauri G, Bertolazzi P, Messa C, Gilardi MC, Chiaradonna F, Vanoni M. Cancer cell growth and survival as a system-level property sustained by enhanced glycolysis and mitochondrial metabolic remodeling. Front Physiol 2012; 3:362. [PMID: 22988443 PMCID: PMC3440026 DOI: 10.3389/fphys.2012.00362] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 08/23/2012] [Indexed: 12/14/2022] Open
Abstract
Systems Biology holds that complex cellular functions are generated as system-level properties endowed with robustness, each involving large networks of molecular determinants, generally identified by “omics” analyses. In this paper we describe four basic cancer cell properties that can easily be investigated in vitro: enhanced proliferation, evasion from apoptosis, genomic instability, and inability to undergo oncogene-induced senescence. Focusing our analysis on a K-ras dependent transformation system, we show that enhanced proliferation and evasion from apoptosis are closely linked, and present findings that indicate how a large metabolic remodeling sustains the enhanced growth ability. Network analysis of transcriptional profiling gives the first indication on this remodeling, further supported by biochemical investigations and metabolic flux analysis (MFA). Enhanced glycolysis, down-regulation of TCA cycle, decoupling of glucose and glutamine utilization, with increased reductive carboxylation of glutamine, so to yield a sustained production of growth building blocks and glutathione, are the hallmarks of enhanced proliferation. Low glucose availability specifically induces cell death in K-ras transformed cells, while PKA activation reverts this effect, possibly through at least two mitochondrial targets. The central role of mitochondria in determining the two investigated cancer cell properties is finally discussed. Taken together the findings reported herein indicate that a system-level property is sustained by a cascade of interconnected biochemical pathways that behave differently in normal and in transformed cells.
Collapse
Affiliation(s)
- Lilia Alberghina
- SysBio Centre for Systems Biology Milano and Rome, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Soundararajan A, Abraham J, Nelon LD, Prajapati SI, Zarzabal LA, Michalek JE, McHardy SF, Hawkins DS, Malempati S, Keller C. 18F-FDG microPET imaging detects early transient response to an IGF1R inhibitor in genetically engineered rhabdomyosarcoma models. Pediatr Blood Cancer 2012; 59:485-92. [PMID: 22238194 PMCID: PMC3924883 DOI: 10.1002/pbc.24075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 12/16/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND Alveolar rhabdomyosarcoma (ARMS) and embryonal rhabdomyosarcoma (ERMS) are among the most common and most treatment resistant soft tissue sarcomas of childhood. Here, we evaluated the potential of (18)F-Fluorodeoxyglucose (FDG) as a marker of therapeutic response to picropodophyllin (PPP), an IGF1R inhibitor, in a conditional mouse model of ARMS and a conditional model of ERMS/undifferentiated pleomorphic sarcoma (UPS). PROCEDURE Primary tumor cell cultures from Myf6Cre,Pax3:Fkhr,p53 and Pax7CreER,Ptch1,p53 conditional models of ARMS and ERMS/UPS were found to be highly sensitive to PPP (IC(50) values 150 and 200 nM, respectively). Animals of each model were then treated with 80 mg/kg/day PPP by intraperitoneal injection for 12 days and imaged by (18)F-FDG microPET. RESULTS Tumor volumes on day 4 for PPP-treated ARMS and ERMS mice were lower than untreated control mouse tumor volumes, although treated tumors were larger than day 0. However, tumor FDG uptake was significantly reduced on day 4 for PPP-treated mice compared to pretreatment baseline or untreated control mice on day 4 (P < 0.05). Nevertheless, by day 12 tumor volumes and FDG uptake for treated mice had increased significantly, indicating rapidly evolving resistance to therapy. CONCLUSIONS (18)F-FDG PET imaging is a potential imaging biomarker of molecular susceptibility to targeted agents early in treatment for this aggressive form of sarcoma, but may find best use serially for Phase I/II studies where chemotherapy and targeted agents are combined to cytoreduce tumors and abrogate Igf1r inhibitor resistance.
Collapse
Affiliation(s)
- Anuradha Soundararajan
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229 USA
| | - Jinu Abraham
- Pediatric Cancer Biology Program, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA
| | - Laura D. Nelon
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229 USA
| | - Suresh I. Prajapati
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229 USA
| | - Lee Ann Zarzabal
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, TX 78229 USA
| | - Joel E. Michalek
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, TX 78229 USA
| | | | - Douglas S. Hawkins
- Division of Hematology/Oncology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, Washington
| | - Suman Malempati
- Division of Pediatric Hematology-Oncology, Oregon Health & Science University, Portland, OR 97239 USA
| | - Charles Keller
- Pediatric Cancer Biology Program, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA,corresponding author: Pediatric Cancer Biology Program, Pape’ Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Mail Code: L321, Portland, OR 97239-3098, Tel 503.494.1210, Fax 503.418.5044,
| |
Collapse
|
21
|
Grabellus F, Worm K, Schmid KW, Sheu SY. The BRAF V600E mutation in papillary thyroid carcinoma is associated with glucose transporter 1 overexpression. Thyroid 2012; 22:377-82. [PMID: 22376167 DOI: 10.1089/thy.2011.0401] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND The glucose transporter 1 (GLUT1) is a key protein that facilitates the extensive glucose uptake of cancer cells, and its overexpression is associated with more aggressive tumor phenotypes. In cases of BRAF mutations, GLUT1 seems to be a target of the constitutive activation of the RAF/MEK/ERK pathway. In this study, we hypothesized that the common BRAF V600E mutation was associated with GLUT1 overexpression and proliferation in papillary thyroid carcinomas (PTCs). METHODS A total of 57 cases of paraffin-embedded PTC (31 BRAF V600E, 26 wild-type BRAF) were investigated using immunohistochemistry with antibodies against GLUT1 and Ki-67 (MK167) protein. The BRAF V600E mutations were detected using direct sequencing of genomic DNA that was isolated from formalin-fixed paraffin-embedded tumor tissues. GLUT1 expression was assessed using the Remmele immunoreactive score and subdivided into three groups (I=negative, II=weakly positive, and III=positive). The Ki-67 labeling index (Ki-67 LI) was determined by counting Ki-67-positive nuclei. RESULTS GLUT1 expression was found in 39/57 (68.4%) samples of PTC. The occurrence of the BRAF V600E genetic variant was significantly correlated with GLUT1 overexpression (p=0.007) and showed a trend toward higher proliferation, which was indicated by Ki-67 LI (p=0.06). Moreover, GLUT1 overexpression was positively associated with Ki-67 labeling (p=0.023). CONCLUSIONS The V600E BRAF mutation in PTC may contribute to the initiation of the glycolytic phenotype and confers growth advantages in cancer cells. Better understanding of the molecular mechanisms of cancer cell energy metabolism may lead to the implementation of targeted treatment modalities, which regulate cancer glucose uptake.
Collapse
Affiliation(s)
- Florian Grabellus
- Institute of Pathology and Neuropathology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany.
| | | | | | | |
Collapse
|
22
|
Co-expression of monocarboxylate transporter 1 (MCT1) and its chaperone (CD147) is associated with low survival in patients with gastrointestinal stromal tumors (GISTs). J Bioenerg Biomembr 2012; 44:171-8. [PMID: 22281667 DOI: 10.1007/s10863-012-9408-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 12/27/2011] [Indexed: 12/19/2022]
Abstract
Monocarboxylate transporters (MCTs) have been described to play an important role in cancer, but to date there are no reports on the significance of MCT expression in gastrointestinal stromal tumors (GISTs). The aim of the present work was to assess the value of MCT expression, as well as co-expression with the MCT chaperone CD147 in GISTs and evaluate their clinical-pathological significance. We analyzed the immunohistochemical expression of MCT1, MCT2, MCT4 and CD147 in a series of 64 GISTs molecularly characterized for KIT, PDGFRA and BRAF mutations. MCT1, MCT2 and MCT4 were highly expressed in GISTs. CD147 expression was associated with mutated KIT (p = 0.039), as well as a progressive increase in Fletcher's Risk of Malignancy (p = 0.020). Importantly, co-expression of MCT1 with CD147 was associated with low patient's overall survival (p = 0.037). These findings suggest that co-expression of MCT1 with its chaperone CD147 is involved in GISTs aggressiveness, pointing to a contribution of cancer cell metabolic adaptations in GIST development and/or progression.
Collapse
|
23
|
Novello S, Giaj Levra M, Vavalà T. Functional imaging in predicting response to antineoplastic agents and molecular targeted therapies in lung cancer: a review of existing evidence. Crit Rev Oncol Hematol 2011; 83:208-15. [PMID: 22062925 DOI: 10.1016/j.critrevonc.2011.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/18/2011] [Accepted: 09/28/2011] [Indexed: 11/17/2022] Open
Abstract
The increasing use of FDG-PET ((18)F-2-fluoro-2-deoxy-d-glucose positron emission tomography) imaging in the staging of non-small-cell lung cancer (NSCLC) may result in a significant shift in stage distribution, with an increased percentage of patients staged as having metastatic disease and consequently a higher percentage of patients treated with systemic therapy. The amount of FDG-PET uptake in primary lung lesions has been shown to be correlated with tumour growth rate. Data suggest that tumours with increased glucose uptake are presumably more metabolically active and more biologically aggressive, and standardized uptake value (SUV) at PET may be regarded as a prognostic factor. Growing evidence suggests that PET may be used as a predictive marker to assess the activity of antineoplastic agents, allowing close monitoring of the efficacy of the treatment in order to be able to switch earlier to alternative therapies according to the individual chemosensitivity of the tumour. Currently the value of FDG-PET for monitoring response is complicated by the heterogeneity of the published data on the methods used for FDG quantification and the selection of the primary targets and clinical endpoints. As a result, objective validation of proposed thresholds of responsiveness is lacking. This article discusses the assessment of treatment response in NSCLC patients using functional imaging, and emphasizes advantages and limitations in clinical management.
Collapse
Affiliation(s)
- S Novello
- Thoracic Oncology Unit, Department of Clinical & Biological Sciences, University of Turin, AOU San Luigi Orbassano, Italy.
| | | | | |
Collapse
|
24
|
Animal tumor models for PET in drug development. Ann Nucl Med 2011; 25:717-31. [DOI: 10.1007/s12149-011-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
25
|
Chiaradonna F, Moresco RM, Airoldi C, Gaglio D, Palorini R, Nicotra F, Messa C, Alberghina L. From cancer metabolism to new biomarkers and drug targets. Biotechnol Adv 2011; 30:30-51. [PMID: 21802503 DOI: 10.1016/j.biotechadv.2011.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/13/2011] [Indexed: 12/14/2022]
Abstract
Great interest is presently given to the analysis of metabolic changes that take place specifically in cancer cells. In this review we summarize the alterations in glycolysis, glutamine utilization, fatty acid synthesis and mitochondrial function that have been reported to occur in cancer cells and in human tumors. We then propose considering cancer as a system-level disease and argue how two hallmarks of cancer, enhanced cell proliferation and evasion from apoptosis, may be evaluated as system-level properties, and how this perspective is going to modify drug discovery. Given the relevance of the analysis of metabolism both for studies on the molecular basis of cancer cell phenotype and for clinical applications, the more relevant technologies for this purpose, from metabolome and metabolic flux analysis in cells by Nuclear Magnetic Resonance and Mass Spectrometry technologies to positron emission tomography on patients, are analyzed. The perspectives offered by specific changes in metabolism for a new drug discovery strategy for cancer are discussed and a survey of the industrial activity already going on in the field is reported.
Collapse
Affiliation(s)
- F Chiaradonna
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Nannini M, Biasco G, Maleddu A, Pantaleo MA. New molecular targets beyond KIT and PDGFRA in gastrointestinal stromal tumors: present and future. Expert Opin Ther Targets 2011; 15:803-15. [DOI: 10.1517/14728222.2011.566215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
27
|
Kinross KM, Brown DV, Kleinschmidt M, Jackson S, Christensen J, Cullinane C, Hicks RJ, Johnstone RW, McArthur GA. In Vivo Activity of Combined PI3K/mTOR and MEK Inhibition in a KrasG12D;Pten Deletion Mouse Model of Ovarian Cancer. Mol Cancer Ther 2011; 10:1440-9. [DOI: 10.1158/1535-7163.mct-11-0240] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Tseng JR, Stuart D, Aardalen K, Kaplan A, Aziz N, Hughes NP, Gambhir SS. Use of DNA microarray and small animal positron emission tomography in preclinical drug evaluation of RAF265, a novel B-Raf/VEGFR-2 inhibitor. Neoplasia 2011; 13:266-75. [PMID: 21390189 PMCID: PMC3050869 DOI: 10.1593/neo.101466] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/06/2010] [Accepted: 12/08/2010] [Indexed: 01/22/2023]
Abstract
Positron emission tomography (PET) imaging has become a useful tool for assessing early biologic response to cancer therapy and may be particularly useful in the development of new cancer therapeutics. RAF265, a novel B-Raf/vascular endothelial growth factor receptor-2 inhibitor, was evaluated in the preclinical setting for its ability to inhibit the uptake of PET tracers in the A375M(B-Raf(V600E)) human melanoma cell line. RAF265 inhibited 2-deoxy-2-[(18)F]fluoro-d-glucose (FDG) accumulation in cell culture at 28 hours in a dose-dependent manner. RAF265 also inhibited FDG accumulation in tumor xenografts after 1 day of drug treatment. This decrease persisted for the remaining 2 weeks of treatment. DNA microarray analysis of treated tumor xenografts revealed significantly decreased expression of genes regulating glucose and thymidine metabolism and revealed changes in apoptotic genes, suggesting that the imaging tracers FDG, 3-deoxy-3-[(18)F]fluorothymidine, and annexin V could serve as potential imaging biomarkers for RAF265 therapy monitoring. We concluded that RAF265 is highly efficacious in this xenograft model of human melanoma and decreases glucose metabolism as measured by DNA microarray analysis, cell culture assays, and small animal FDG PET scans as early as 1 day after treatment. Our results support the use of FDG PET in clinical trials with RAF265 to assess early tumor response. DNA microarray analysis and small animal PET studies may be used as complementary technologies in drug development. DNA microarray analysis allows for analysis of drug effects on multiple pathways linked to cancer and can suggest corresponding imaging tracers for further analysis as biomarkers of tumor response.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Drug Evaluation, Preclinical
- Enzyme Inhibitors/therapeutic use
- Female
- Fluorodeoxyglucose F18
- Gene Expression Profiling
- Glucose/metabolism
- Humans
- Imidazoles/therapeutic use
- Immunoenzyme Techniques
- Leukemia, Myeloid, Acute/diagnostic imaging
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Melanoma/diagnostic imaging
- Melanoma/drug therapy
- Melanoma/pathology
- Mice
- Mice, Nude
- Oligonucleotide Array Sequence Analysis
- Proto-Oncogene Proteins B-raf/antagonists & inhibitors
- Pyridines/therapeutic use
- RNA, Messenger/genetics
- Radionuclide Imaging
- Radiopharmaceuticals
- Reverse Transcriptase Polymerase Chain Reaction
- Thymidine/metabolism
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jeffrey R Tseng
- Molecular Imaging Program at Stanford, Bio-X Program, Department of Radiology, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Mileshkin L, Hicks RJ, Hughes BGM, Mitchell PLR, Charu V, Gitlitz BJ, Macfarlane D, Solomon B, Amler LC, Yu W, Pirzkall A, Fine BM. Changes in 18F-fluorodeoxyglucose and 18F-fluorodeoxythymidine positron emission tomography imaging in patients with non-small cell lung cancer treated with erlotinib. Clin Cancer Res 2011; 17:3304-15. [PMID: 21364032 DOI: 10.1158/1078-0432.ccr-10-2763] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE Assessing clinical activity of molecularly targeted anticancer agents, especially in the absence of tumor shrinkage, is challenging. To evaluate on-treatment 18F-fluorodeoxyglucose (FDG) and/or 18F-fluorodeoxythymidine (FLT) positron emission tomography (PET) for this purpose, we conducted a prospective multicenter trial assessing PET response rates and associations with progression-free (PFS) and overall survival (OS) in 2nd/3rd-line non-small-cell lung cancer patients treated with erlotinib. EXPERIMENTAL DESIGN PET/computed tomography (CT) scans were conducted at baseline, day (d)14 and d56 after the first daily erlotinib dose, with diagnostic CT at baseline and d56 (all scans centrally reviewed). PET partial metabolic response (PMR) was defined as a mean decrease (in ≤ 5 lesions/patient) of 15% or more maximum standardized uptake value. PFS was investigator-determined. RESULTS Of 74 erlotinib-treated patients, 51 completed all imaging assessments through d56; 13 of 51 (26%) FDG-evaluable patients had PMR at d14, as did 9 of 50 (18%) FLT-evaluable patients. Four (7.8%) showed partial responses (PR) by d56 CT; all 4 had PMR by d14 FDG-PET with 3 PMRs by d14 FLT-PET. Three of the 4 patients with CT PR had evaluable archival tumor tissue; all 3 had epidermal growth factor receptor mutations. D14 and d56 PMRs by FDG or FLT were associated with improved PFS; HRs for PET responders versus nonresponders were 0.3 to 0.4. D14 FDG-PET PMR was associated with improved OS (P = 0.03) compared with FDG-PET nonresponders. CONCLUSION Early (d14) FDG-PET PMR is associated with improved PFS and OS, even in the absence of subsequent Response Evaluation Criteria in Solid Tumors response. These data support inclusion of FDG-PET imaging in clinical trials testing novel targeted therapies, particularly those with anticipated cytostatic effects.
Collapse
Affiliation(s)
- Linda Mileshkin
- Peter MacCallum Cancer Centre, East Melbourne, The University of Melbourne, Parkville, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
High-throughput small animal PET imaging in cancer research: evaluation of the capability of the Inveon scanner to image four mice simultaneously. Nucl Med Commun 2010; 31:851-8. [PMID: 20683363 DOI: 10.1097/mnm.0b013e32833dc61d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The aim of this study was to assess the capability of small animal PET (SA-PET) devices to image four mice simultaneously to improve the throughput of SA-PET experiments in cancer research. A customized bed was designed to image up to four mice simultaneously. This bed can easily replace the bed provided by the manufacturer and is connected to an anaesthesia device. A mouse-sized phantom was imaged, mimicking simultaneous imaging of four mice with computation of recovery coefficients and spillover ratios (SORs). In addition, eight mice bearing subcutaneous tumours (human embryonal carcinoma, n=22 tumours) were simultaneously imaged in groups of four on an Inveon SA-PET scanner after injection of F-fluoro-D-glucose. Tumour activity (Bq/ml), as determined by the SA-PET, was compared with ex-vivo counting. For a 5-mm rod, recovery coefficients were 1.15 and 1.05 for a phantom imaged at the central field of view or off-centred on the customized bed, respectively. SORair and SORwater were 0.05 and 0.04 for a phantom imaged alone and 0.15 and 0.06 for a phantom imaged with three additional scatter sources, respectively. Correlation between SA-PET and ex-vivo quantification was good (r=0.91, P<0.0001). The mean ratio of PET quantitative data and ex-vivo counting was equal to 0.9 (95% confidence interval: 0.70-1.09). New generation SA-PET may be suitable for simultaneously imaging four tumour-bearing mice, although improvement in scatter correction efficiency appears necessary. The type of customized bed developed in this study could be easily adapted to other large-bore SA-PET scanners.
Collapse
|
31
|
|
32
|
Egawa-Takata T, Endo H, Fujita M, Ueda Y, Miyatake T, Okuyama H, Yoshino K, Kamiura S, Enomoto T, Kimura T, Inoue M. Early reduction of glucose uptake after cisplatin treatment is a marker of cisplatin sensitivity in ovarian cancer. Cancer Sci 2010; 101:2171-8. [PMID: 20678156 PMCID: PMC11158957 DOI: 10.1111/j.1349-7006.2010.01670.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cisplatin is an effective chemotherapeutic agent for ovarian cancer, but the sensitivity of cancers differs in individual cases. Because cisplatin is reported to suppress glucose uptake, we investigated the correlation between glucose uptake and sensitivity to the drug. A fluorescent derivative of D-glucose, 2-NBDG (2-[N-(7-nitrobenz-2-oxa-1,3-diaxol-4-yl) amino]-2-deoxyglucose), was used to evaluate glucose uptake. Two ovarian cancer cell lines, SKOV-3 as a relatively resistant line and OVCAR-3 as a relatively sensitive line, were analyzed. Both cell lines had a decreased number of cells accompanied by cell death 24 h after cisplatin treatment, but not at 3 h. In contrast, glucose uptake was decreased 3 h after high-dose cisplatin treatment, which correlated with the sensitivity to the drug at 24 h. The protein levels of glucose transporter 1 (GLUT1) did not change with cisplatin treatment. In contrast, the membrane localization of GLUT1 disappeared after cisplatin treatment. Other cisplatin-resistant cell lines did not show an early decrease in glucose uptake after cisplatin treatment. The early decrease in glucose uptake and later cell death also correlated in cultured cancer cells from ovarian cancer patients. Thus, the decrease in glucose uptake at an early time point after high dose cisplatin treatment reflected cisplatin chemosensitivity in ovarian cancer cells. Measuring glucose uptake might be useful as a rapid evaluation of cisplatin chemosensitivity in ovarian cancer patients.
Collapse
Affiliation(s)
- Tomomi Egawa-Takata
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vergez S, Delord JP, Thomas F, Rochaix P, Caselles O, Filleron T, Brillouet S, Canal P, Courbon F, Allal BC. Preclinical and clinical evidence that Deoxy-2-[18F]fluoro-D-glucose positron emission tomography with computed tomography is a reliable tool for the detection of early molecular responses to erlotinib in head and neck cancer. Clin Cancer Res 2010; 16:4434-45. [PMID: 20660574 DOI: 10.1158/1078-0432.ccr-09-2795] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE There is a clinical need to identify predictive markers of the responses to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKI). Deoxy-2-[(18)F]fluoro-d-glucose positron emission tomography with computed tomography ((18)FDG-PET/CT) could be a tool of choice for monitoring the early effects of this class of agent on tumor activity. EXPERIMENTAL DESIGN Using models of human head and neck carcinoma (CAL33 and CAL166 cell lines), we first tested in vitro and in vivo whether the in vivo changes in (18)FDG-PET/CT uptake were associated with the molecular and cellular effects of the EGFR-TKI erlotinib. Then, the pathologic and morphologic changes and the (18)FDG-PET/CT uptake before and after erlotinib exposure in patients were analyzed. RESULTS Erlotinib strongly inhibited extracellular signal-regulated kinase-1/2 (ERK-1/2) phosphorylation both in the preclinical models and in patients. Western blotting, immunofluorescence, and immunohistochemistry showed that erlotinib did not modify Glut-1 expression at the protein level either in cell line models or in tumor tissue from mouse xenografts or in patients. Phospho-ERK-1/2 inhibition was associated with a reduction in (18)FDG uptake in animal and human tumors. The biological volume was more accurate than the standardized uptake value for the evaluation of the molecular responses. CONCLUSION These results show that the (18)FDG-PET/CT response is a reliable surrogate marker of the effects of erlotinib in head and neck carcinoma.
Collapse
Affiliation(s)
- Sébastien Vergez
- Université Paul Sabatier and Institut Claudius Regaud, Toulouse Cedex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cullinane C, Natoli A, Hui Y, Conus N, Jackson S, Brüggen J, Manley PW, McArthur GA. Preclinical evaluation of nilotinib efficacy in an imatinib-resistant KIT-driven tumor model. Mol Cancer Ther 2010; 9:1461-8. [PMID: 20442311 DOI: 10.1158/1535-7163.mct-09-1181] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The novel KIT inhibitor nilotinib is currently being evaluated for its clinical utility in the treatment of gastrointestinal stromal tumor. However, the effects of nilotinib in cells expressing commonly occurring KIT mutations remain to be fully defined. The aim of this study was therefore to investigate the efficacy of nilotinib against cells expressing imatinib-sensitive or imatinib-resistant KIT mutations and to evaluate [(18)F] fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging as a biomarker of nilotinib response in vivo. Nilotinib inhibited the proliferation of imatinib-responsive V560G-KIT FDC-P1 and imatinib-resistant D816V-KIT FDC-P1 cells with a GI(50) of 4.9 and 630 nmol/L, respectively, whereas apoptosis studies revealed that nilotinib and imatinib were equipotent against the V560G cell line. In contrast, although 10 micromol/L nilotinib induced >50% apoptosis in the D816V cells at 16 hours, 10 micromol/L imatinib had no effect on cell survival at 24 hours. Syngeneic DBA2/J mice bearing FDC-P1-KIT tumors were evaluated for response to nilotinib by FDG-PET. V560G-KIT FDC-P1 tumor FDG uptake was significantly reduced compared with baseline levels following 2 days of nilotinib treatment. In contrast, no effect of nilotinib was observed on tumor growth or FDG-PET uptake into D816V tumors despite intratumoral drug levels reaching in excess of 10 micromol/L at 4 hours after dosing. Biomarker analysis revealed the inhibition of KIT phosphorylation in V560G but not D816V tumors. These findings show the in vivo activity of nilotinib in the treatment of tumors bearing V560G-KIT but not D816V-KIT and the utility of FDG-PET imaging to assess tumor response to this agent.
Collapse
Affiliation(s)
- Carleen Cullinane
- Translational Research Laboratory, Research Division, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Pan G, Winter TN, Roberts JC, Fairbanks CA, Elmquist WF. Organic cation uptake is enhanced in bcrp1-transfected MDCKII cells. Mol Pharm 2010; 7:138-45. [PMID: 19886673 DOI: 10.1021/mp900177r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stably transfected cell models are routinely used to examine drug-transporter interactions. In one such model of bcrp1-transfected MDCKII cells, we observed a significant enhancement of organic cation intracellular accumulation. Therefore, our goal was to further explore the expression and functional consequences of this cation transport system. Transport assays were carried out in wild-type and bcrp1-transfected MDCKII cells to examine uptake of [3H]-prazosin (bcrp1 positive control), [3H]-agmatine, [3H]-TEA, and [14C]-choline. RT-PCR was employed to determine the mRNA levels of bcrp1 and OCT2/OCT3. Western blots were used to evaluate corresponding protein levels. Accumulation studies determined a significant increase in the uptake of the organic cations agmatine, TEA, and choline in bcrp1-transfected cells when compared to wild-type cells. Directional transport of [3H]-agmatine showed a significantly greater apical (A) to basolateral (B) than B-to-A flux in both cell types. In spite of this, the A-to-B flux was significantly lower in bcrp1-transfected cells. RT-PCR revealed 10-fold higher OCT2 mRNA levels in bcrp1-transfected cells, with no changes in OCT3. OCT2 protein expression was approximately 3.5-fold higher in bcrp1-transfected cells. The upregulation of OCT2 in bcrp1-transfected MDCKII cells contributed to a significant enhancement in the uptake of several organic cations. These results are consistent with the endogenous expression of OCT2 in the kidney tubule, and may be related to the expression and function of bcrp1. Our findings illustrate the importance of understanding how endogenous transporters, which may compete for common substrates, may be influenced by the overexpression and enhanced function of recombinant transport systems.
Collapse
Affiliation(s)
- Guoyu Pan
- Department of Pharmaceutics, University of Minnesota, 308 Harvard Street SE, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
36
|
Passarella RJ, Zhou L, Phillips JG, Wu H, Hallahan DE, Diaz R. Recombinant peptides as biomarkers for tumor response to molecular targeted therapy. Clin Cancer Res 2009; 15:6421-9. [PMID: 19825959 DOI: 10.1158/1078-0432.ccr-09-0945] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Phage display technology can be used to identify peptide sequences that bind rapidly and specifically to tumors responding to sunitinib therapy. These peptides may help to address problems with current methods of assessing tumor response to therapy that can be slow and have limited usage. EXPERIMENTAL DESIGN The peptide of interest was isolated after four rounds of biopanning in MDA-MB-231 and MCF-7 xenografted tumors. The binding location of the peptide was investigated with immunohistochemistry. Its in vivo ability to bind to breast tumors responding to therapy was determined by treating nude mice, xenografted with various tumor cell lines, with sunitinib and using near IR imaging to assess the ability of the peptide conjugated to Alexafluor-750 to bind tumors. RESULTS EGEVGLG was the dominant sequence isolated from biopanning. This peptide showed increased binding relative to control groups in two cancer cell lines (MDA-MB-435 and MCF-7 human breast) responding to sunitinib treatment, whereas no elevated binding occurred in vitro when samples were incubated with tumor cells that are unresponsive to sunitinib treatment (B16 melanoma and BxPC3 pancreatic). Mice xenografted with tumors that are responsive to sunitinib therapy showed increased peptide binding when compared with untreated control. Mice bearing tumors unresponsive to sunitinib therapy showed no increased peptide binding between treated and untreated groups. CONCLUSION The use of recombinant peptides to assess the pharmacodynamic response of cancer holds promise in minimizing the duration of ineffective treatment regimens in patients, potentially providing a more rapid and less invasive assessment of cancer response to systemic therapy.
Collapse
Affiliation(s)
- Ralph J Passarella
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | |
Collapse
|
37
|
Herrmann K, Krause BJ, Bundschuh RA, Dechow T, Schwaiger M. Monitoring response to therapeutic interventions in patients with cancer. Semin Nucl Med 2009; 39:210-32. [PMID: 19341841 DOI: 10.1053/j.semnuclmed.2008.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Positron emission tomography (PET) and PET/computed tomography (CT) with the glucose analog (18)F-fluorodeoxyglucose (FDG) are increasingly used to assess response to therapy in patients, and there is converging evidence that changes in glucose utilization during therapy can be used to predict clinical outcome. Today, integrated PET/CT systems have mainly replaced stand-alone PET devices, providing the opportunity to integrate morphologic information and functional information. However, the use of PET/CT systems also gives rise to methodological challenges for the quantitative analysis of PET scans for treatment monitoring. Recently published single-center studies demonstrate that FDG-PET and FDG-PET/CT have been successfully used for monitoring of tumor response to cytotoxic therapy in a variety of tumor entities. The potential early identification of nonresponding tumors provides an opportunity to alter treatment regimens according to the individual chemosensitivity of the tumor tissue. In this article, we review the methodological background to monitoring of cancer treatment with PET/CT, the diagnostic and prognostic performance of PET/CT for predicting tumor response with the glucose analog FDG in various tumor entities, and the clinical potential of new imaging probes. In addition, the future direction of research and clinical applications is discussed.
Collapse
Affiliation(s)
- Ken Herrmann
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany.
| | | | | | | | | |
Collapse
|
38
|
Monazzam A, Razifar P, Ide S, Rugaard Jensen M, Josephsson R, Blomqvist C, Langström B, Bergström M. Evaluation of the Hsp90 inhibitor NVP-AUY922 in multicellular tumour spheroids with respect to effects on growth and PET tracer uptake. Nucl Med Biol 2009; 36:335-42. [PMID: 19324279 DOI: 10.1016/j.nucmedbio.2008.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 12/08/2008] [Accepted: 12/24/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Molecular targeting has become a prominent concept in cancer treatment and heat shock protein 90 (Hsp90) inhibitors are suggested as promising anticancer drugs. The Hsp90 complex is one of the chaperones that facilitate the refolding of unfolded or misfolded proteins and plays a role for key oncogenic proteins such as Her2, Raf-1, Akt/PKB, and mutant p53. NVP-AUY922 is a novel low-molecular Hsp90 inhibitor, currently under clinical development as an anticancer drug. Disruption of the Hsp90-client protein complexes leads to proteasome-mediated degradation of client proteins and cell death. The aim of the current study was to use a combination of the multicellular tumour spheroid (MTS) model and positron emission tomography (PET) to investigate the effects of NVP-AUY922 on tumour growth and its relation to PET tracer uptake for the selection of appropriate PET tracer. A further aim was to evaluate the concentration and time dependence in the relation between growth inhibition and PET tracer uptake as part of translational imaging activities. METHODS MTS of two breast cancer cell lines (MCF-7 and BT474), one glioblastoma cell line (U87MG) and one colon carcinoma cell line (HCT116) were prepared. Initially, we investigated MTS growth pattern and (3)H-thymidine incorporation in MTS after continuous exposure to NVP-AUY922 in order to determine dose response. Then the short-term effect of the drug on the four PET tracers 2-[(18)F] fluoro-2-deoxyglucose (FDG), 3'-deoxy-3'-fluorothymidine (FLT), methionine and choline was correlated to the long-term effect (changes in growth pattern) to determine the adequate PET tracer with high predictability. Next, the growth inhibitory effect of different dose schedules was evaluated to determine the optimal dose and time. Finally, the effect of a 2-h exposure to the drug on growth pattern and FDG/FLT uptake was evaluated. RESULTS A dose-dependent inhibition of growth and decrease of (3)H-thymidine uptake was observed with 100% growth cessation in the dose range 7-52 nM and 50% (3)H-thymidine reduction in the range of 10-23 nM, with the most pronounced effect on BT474 cells. The effect of the drug was best detected by FLT. The results suggested that a complete cessation of growth of the viable cell volume was achieved with about 50% inhibition of FLT uptake 3 days after continuous treatment. Significant growth inhibition was observed at all doses and all exposure time spans. Two-hour exposure to NVP-AUY922 generated a growth inhibition which persisted dose dependently up to 10 days. The uptake of FDG per viable tumour volume was reduced by just 25% with 300 nM treatment of the drug, whereas the FLT uptake decreased up to 75% in correlation with the growth inhibition and recovery. CONCLUSIONS Our results indicate a prolonged action of NVP-AUY922 in this cell culture, FLT is a suitable tracer for the monitoring of the effect and a FLT PET study within 3 days after treatment can predict the treatment outcome in this model. If relevant in vivo, this information can be used for efficient planning of animal PET studies and later human PET trial.
Collapse
Affiliation(s)
- Azita Monazzam
- Institute of Oncology, Radiology and Clinical Immunology, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Janeway KA, Albritton KH, Van Den Abbeele AD, D'Amato GZ, Pedrazzoli P, Siena S, Picus J, Butrynski JE, Schlemmer M, Heinrich MC, Demetri GD. Sunitinib treatment in pediatric patients with advanced GIST following failure of imatinib. Pediatr Blood Cancer 2009; 52:767-71. [PMID: 19326424 DOI: 10.1002/pbc.21909] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sunitinib inhibits KIT and other members of the split-kinase-domain family of receptor tyrosine kinases. Sunitinib prolongs survival in adult patients with imatinib-resistant gastrointestinal stromal tumor (GIST). We report the experience with sunitinib in pediatric patients with advanced GIST following failure of imatinib. PROCEDURE Sunitinib therapy was provided through a treatment-use protocol. Patients were 10-17 years old at enrollment. All patients had GIST resistant to imatinib therapy. Sunitinib was administered daily for 4 weeks in 6-week treatment cycles. KIT and platelet-derived growth factor receptor alpha (PDGFRA) genotyping of tumor tissue were performed. RESULTS One patient achieved a partial response, five patients had stable disease and one patient had progressive disease on sunitinib. The duration of disease stabilization was between 7 and 21+ months, with a mean of 15 months. Time to tumor progression was longer on sunitinib than on prior imatinib treatment for five of six patients. Two patients experienced grade 3 adverse events. All other adverse events were grade 1-2. None of the five patients tested had mutations in KIT or PDGFRA. CONCLUSION Sunitinib treatment was associated with substantial initial antitumor activity and acceptable tolerability in this group of pediatric patients with imatinib-resistant GIST.
Collapse
Affiliation(s)
- Katherine A Janeway
- Pediatric Oncology, Dana-Farber Cancer Institute and Department of Medicine, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The definition of molecular imaging provided by the Society of Nuclear Medicine is "the visualization, characterization and measurement of biological processes at the molecular and cellular levels in humans and other living systems". This review gives an overview of the technologies available for and the potential benefits from molecular imaging at the preclinical stage. It focuses on the use of imaging probes based on bioconjugates and for reasons of brevity confines itself to discussion of applications in the field of oncology, although molecular imaging can be equally useful in many fields including cardiovascular medicine, neurosciences, infection, and others.
Collapse
Affiliation(s)
- Stephen Mather
- Barts and The London Queen Mary's School of Medicine and Dentistry, Centre for Cancer Imaging Institute of Cancer and the CR-UK Clinical Centre, St. Bartholomew's Hos, United Kingdon.
| |
Collapse
|
41
|
Noninvasive structural, functional, and molecular imaging in drug development. Curr Opin Chem Biol 2009; 13:360-71. [DOI: 10.1016/j.cbpa.2009.03.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/30/2009] [Indexed: 11/19/2022]
|
42
|
In Vivo Measurements of Tumor Metabolism and Growth after Administration of Enzastaurin Using Small Animal FDG Positron Emission Tomography. JOURNAL OF ONCOLOGY 2009; 2009:596560. [PMID: 19503801 PMCID: PMC2688651 DOI: 10.1155/2009/596560] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 03/13/2009] [Indexed: 12/20/2022]
Abstract
Background. The use of 2-[(18)F]fluoro-2-deoxy-D-glucose ([(18)F]FDG) may help to establish the antitumor activity of enzastaurin, a novel protein kinase C-beta II (PKC-betaII) inhibitor, in mouse xenografts. Methods. The hematologic cell line RAJI and the solid tumor cell line U87MG were each implanted in NOD/SCID mice. Standard tumor growth measurements and [(18)F]FDG PET imaging were performed weekly for up to three weeks after tumor implantation and growth. Results. Concomitant with caliper measurements, [(18)F]FDG PET imaging was performed to monitor glucose metabolism. Heterogeneity of glucose uptake in various areas of the tumors was observed after vehicle or enzastaurin treatment. This heterogeneity may limit the use of [(18)F]FDG PET imaging to measure enzastaurin-associated changes in xenograft tumors. Conclusion. [(18)F]FDG PET imaging technique does not correlate with standard caliper assessments in xenografts to assess the antitumor activity of enzastaurin. Future studies are needed to determine the use of [(18)F]FDG PET imaging in preclinical models.
Collapse
|
43
|
Kansara M, Tsang M, Kodjabachian L, Sims NA, Trivett MK, Ehrich M, Dobrovic A, Slavin J, Choong PFM, Simmons PJ, Dawid IB, Thomas DM. Wnt inhibitory factor 1 is epigenetically silenced in human osteosarcoma, and targeted disruption accelerates osteosarcomagenesis in mice. J Clin Invest 2009; 119:837-51. [PMID: 19307728 DOI: 10.1172/jci37175] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 01/21/2009] [Indexed: 02/06/2023] Open
Abstract
Wnt signaling increases bone mass by stimulating osteoblast lineage commitment and expansion and forms the basis for novel anabolic therapeutic strategies being developed for osteoporosis. These strategies include derepression of Wnt signaling by targeting secreted Wnt pathway antagonists, such as sclerostin. However, such therapies are associated with safety concerns regarding an increased risk of osteosarcoma, the most common primary malignancy of bone. Here, we analyzed 5 human osteosarcoma cell lines in a high-throughput screen for epigenetically silenced tumor suppressor genes and identified Wnt inhibitory factor 1 (WIF1), which encodes an endogenous secreted Wnt pathway antagonist, as a candidate tumor suppressor gene. In vitro, WIF1 suppressed beta-catenin levels in human osteosarcoma cell lines, induced differentiation of human and mouse primary osteoblasts, and suppressed the growth of mouse and human osteosarcoma cell lines. Wif1 was highly expressed in the developing and mature mouse skeleton, and, although it was dispensable for normal development, targeted deletion of mouse Wif1 accelerated development of radiation-induced osteosarcomas in vivo. In primary human osteosarcomas, silencing of WIF1 by promoter hypermethylation was associated with loss of differentiation, increased beta-catenin levels, and increased proliferation. These data lead us to suggest that derepression of Wnt signaling by targeting secreted Wnt antagonists in osteoblasts may increase susceptibility to osteosarcoma.
Collapse
Affiliation(s)
- Maya Kansara
- Ian Potter Foundation Centre for Cancer Genetics and Preventative Medicine, and Sir Donald and Lady Trescowthick Laboratories, Peter MacCallumCancer Centre, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pantaleo MA, Landuzzi L, Nicoletti G, Nanni C, Boschi S, Piazzi G, Santini D, Di Battista M, Castellucci P, Lodi F, Fanti S, Lollini PL, Biasco G. Advances in preclinical therapeutics development using small animal imaging and molecular analyses: the gastrointestinal stromal tumors model. Clin Exp Med 2009; 9:199-205. [PMID: 19225718 PMCID: PMC2709232 DOI: 10.1007/s10238-009-0033-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 01/16/2009] [Indexed: 01/13/2023]
Abstract
The large use of target therapies in the treatment of gastrointestinal stromal tumors (GISTs) highlighted the urgency to integrate new molecular imaging technologies, to develop new criteria for tumor response evaluation and to reach a more comprehensive definition of the molecular target. These aspects, which come from clinical experiences, are not considered enough in preclinical research studies which aim to evaluate the efficacy of new drugs or new combination of drugs with molecular target. We developed a xenograft animal model GIST882 using nude mice. We evaluated both the molecular and functional characterization of the tumor mass. The mutational analysis of KIT receptor of the GIST882 cell lines and tumor mass showed a mutation on exon 13 that was still present after in vivo cell growth. The glucose metabolism and cell proliferation was evaluated with a small animal PET using both FDG and FLT. The experimental development of new therapies for GIST treatment requires sophisticated animal models in order to represent the tumor molecular heterogeneity already demonstrated in the clinical setting and in order to evaluate the efficacy of the treatment also considering the inhibition of tumor metabolism, and not only considering the change in size of tumors. This approach of cancer research on GISTs is crucial and essential for innovative perspectives that could cross over to other types of cancer.
Collapse
Affiliation(s)
- M A Pantaleo
- Department of Hematology and Oncology Sciences L.A.Seragnoli, Sant'Orsola-Malpighi Hospital, University of Bologna, 40138, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Prior JO, Montemurro M, Orcurto MV, Michielin O, Luthi F, Benhattar J, Guillou L, Elsig V, Stupp R, Delaloye AB, Leyvraz S. Early prediction of response to sunitinib after imatinib failure by 18F-fluorodeoxyglucose positron emission tomography in patients with gastrointestinal stromal tumor. J Clin Oncol 2008; 27:439-45. [PMID: 19064982 DOI: 10.1200/jco.2008.17.2742] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Positron emission tomography with (18)F-fluorodeoxyglucose (FDG-PET) was used to evaluate treatment response in patients with gastrointestinal stromal tumors (GIST) after administration of sunitinib, a multitargeted tyrosine kinase inhibitor, after imatinib failure. PATIENTS AND METHODS Tumor metabolism was assessed with FDG-PET before and after the first 4 weeks of sunitinib therapy in 23 patients who received one to 12 cycles of sunitinib therapy (4 weeks of 50 mg/d, 2 weeks off). Treatment response was expressed as the percent change in maximal standardized uptake values (SUV). The primary end point of time to tumor progression was compared with early PET results on the basis of traditional Response Evaluation Criteria in Solid Tumors (RECIST) criteria. RESULTS Progression-free survival (PFS) was correlated with early FDG-PET metabolic response (P < .0001). Using -25% and +25% thresholds for SUV variations from baseline, early FDG-PET response was stratified in metabolic partial response, metabolically stable disease, or metabolically progressive disease; median PFS rates were 29, 16, and 4 weeks, respectively. Similarly, when a single FDG-PET positive/negative was considered after 4 weeks of sunitinib, the median PFS was 29 weeks for SUVs less than 8 g/mL versus 4 weeks for SUVs of 8 g/mL or greater (P < .0001). None of the patients with metabolically progressive disease subsequently responded according to RECIST criteria. Multivariate analysis showed shorter PFS in patients who had higher residual SUVs (P < .0001), primary resistance to imatinib (P = .024), or nongastric GIST (P = .002), regardless of the mutational status of the KIT and PDGFRA genes. CONCLUSION Week 4 FDG-PET is useful for early assessment of treatment response and for the prediction of clinical outcome. Thus, it offers opportunities to individualize and optimize patient therapy.
Collapse
Affiliation(s)
- John O Prior
- Nuclear Medicine Department,Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Early evaluation of the effects of chemotherapy with longitudinal FDG small-animal PET in human testicular cancer xenografts: early flare response does not reflect refractory disease. Eur J Nucl Med Mol Imaging 2008; 36:396-405. [PMID: 19050878 DOI: 10.1007/s00259-008-0984-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 10/09/2008] [Indexed: 01/27/2023]
Abstract
AIM We aimed to evaluate the usefulness of FDG PET in the early prediction of the effects of chemotherapy on human testicular cancer xenografts. MATERIAL AND METHODS Nude rats bearing subcutaneous human embryonal carcinoma xenografts received either cisplatin (5 mg/kg) or saline serum. Small-animal PET studies were performed on days 0, 2, 4 and 7 and compared to immunochemistry studies, flow cytometry studies and hexokinase assays. RESULTS Cisplatin treatment resulted in biphasic FDG uptake evolution: a peak was observed on day 2, followed by a marked decrease on day 7 despite an insignificant change in tumour volume. Similarly, a peak in cyclin A immunostaining was observed on days 2 and 4), followed by a significant decrease on day 7. Flow cytometry showed that the cyclin A peak was not related to increased cell proliferation but was due to a transient S and G(2)/M cell cycle arrest. A marked increase in cell apoptosis was observed from day 2 to day 7. GLUT-1 showed a significant decrease on day 7. Macrophagic infiltrate remained stable except for an increase observed on day 7. In control tumours, continuous growth was observed, all immunostaining markers remaining stable over time. Hexokinase activity was significantly lower on day 7 in treated tumours than in controls. CONCLUSION FDG PET may be useful in the early evaluation of treatment in patients with testicular cancer. In our model, a very early increased [(18)F]-FDG uptake was related to a transient cell cycle arrest and early stage apoptosis but did not reveal refractory disease.
Collapse
|
47
|
Comparison of semiquantitative fluorescence imaging and PET tracer uptake in mesothelioma models as a monitoring system for growth and therapeutic effects. Nucl Med Biol 2008; 35:851-60. [DOI: 10.1016/j.nucmedbio.2008.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 08/10/2008] [Accepted: 08/26/2008] [Indexed: 11/22/2022]
|
48
|
Abstract
Molecular imaging of tumor metabolism has gained considerable interest, since preclinical studies have indicated a close relationship between the activation of various oncogenes and alterations of cellular metabolism. Furthermore, several clinical trials have shown that metabolic imaging can significantly impact patient management by improving tumor staging, restaging, radiation treatment planning, and monitoring of tumor response to therapy. In this review, we summarize recent data on the molecular mechanisms underlying the increased metabolic activity of cancer cells and discuss imaging techniques for studies of tumor glucose, lipid, and amino acid metabolism.
Collapse
Affiliation(s)
- Christian Plathow
- Department of Nuclear Medicine, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
49
|
Aide N, Labiche A, Herlin P, Paciencia M, Poulain L, Dutoit S, Montravers F, Gauduchon P, Chasle J. Usefulness of automatic quantification of immunochemical staining on whole tumor sections for correlation with oncological small animal PET studies: an example with cell proliferation, glucose transporter 1 and FDG. Mol Imaging Biol 2008; 10:237-44. [PMID: 18543043 DOI: 10.1007/s11307-008-0144-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 02/14/2008] [Accepted: 03/03/2008] [Indexed: 10/22/2022]
Abstract
AIM To highlight the use of automatic quantification of immunochemical staining on digitized images of whole tumor sections in preclinical positron emission tomography (PET) studies. MATERIALS AND METHODS Xenografted human testicular tumors (36) were imaged with 2-deoxy-2[F-18]fluoro-D: -glucose (FDG) small animal PET (SA-PET). Tumor cell proliferation and glucose transportation were assessed with cyclin A and Glut-1 immunostaining. Tumor slides were digitized and processed with PixCyt software enabling whole slide quantification, then compared with junior and senior pathologist manual scoring. Manual and automatic quantification results were correlated to FDG uptake. RESULTS For cyclin A, inter- and intra-observer agreement for manual scoring was 0.52 and 0.72 and concordance between senior pathologist and automatic quantification was 0.84. Correlations between Tumor/Background ratio and tumor cell proliferation assessed by automatic quantification, junior and senior pathologists were 0.75, 0.55, and 0.61, respectively. Correlation between Tumor/Background ratio and Glut-1 assessed by automatic quantification was 0.74. CONCLUSION Automatic quantification of immunostaining is a valuable tool to overcome inter- and intra-observer variability for correlation of cell proliferation or other markers with tumor tracer uptake.
Collapse
Affiliation(s)
- Nicolas Aide
- GRECAN, EA 1772, François Baclesse Comprehensive Cancer Center and Caen University, Caen, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
|