1
|
Cangut B, Akinlusi R, Mohseny A, Ghesani N, Ghesani M. Evolving Paradigms in Lung Cancer: Latest Trends in Diagnosis, Management, and Radiopharmaceuticals. Semin Nucl Med 2025; 55:264-276. [PMID: 40055050 DOI: 10.1053/j.semnuclmed.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025]
Abstract
Lung cancer is one of the most common and deadliest forms of cancer worldwide. Over the past two decades, significant changes have occurred in the classification of lung cancer, involving multidisciplinary input and emphasizing the growing contribution of immunohistochemistry and molecular techniques to morphology in the classification scheme. This comprehensive review will cover the background and epidemiology of lung cancer as well as advancements in its staging and management, including discussions of new surgical techniques, targeted therapies, and immunotherapy. The review will detail the role of 18F-FDG-PET-CT in lung cancer, highlighting its importance in staging, treatment response assessment, and recurrence detection. While immunotherapy has transformed lung cancer management and improved patient outcomes, it presents major challenges and opportunities for optimal assessment of treatment response in lung cancer patients using 18F-FDG-PET-CT. This review will also explore future directions, including a discussion of promising new targeted diagnostic radiopharmaceuticals for PET/CT imaging. Additionally, there will be a brief discussion of evolving and exciting treatment options for lung cancer using targeted therapeutic radiopharmaceuticals. Several case-based illustrations are included to exemplify the role of 18F-FDG-PET-CT in various clinical scenarios.
Collapse
Affiliation(s)
- Busra Cangut
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rahman Akinlusi
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ava Mohseny
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nasrin Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Munir Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
2
|
Rajagopal S, Bogaard HJ, Elbaz MSM, Freed BH, Remy-Jardin M, van Beek EJR, Gopalan D, Kiely DG. Emerging multimodality imaging techniques for the pulmonary circulation. Eur Respir J 2024; 64:2401128. [PMID: 39209480 PMCID: PMC11525339 DOI: 10.1183/13993003.01128-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) remains a challenging condition to diagnose, classify and treat. Current approaches to the assessment of PH include echocardiography, ventilation/perfusion scintigraphy, cross-sectional imaging using computed tomography and magnetic resonance imaging, and right heart catheterisation. However, these approaches only provide an indirect readout of the primary pathology of the disease: abnormal vascular remodelling in the pulmonary circulation. With the advent of newer imaging techniques, there is a shift toward increased utilisation of noninvasive high-resolution modalities that offer a more comprehensive cardiopulmonary assessment and improved visualisation of the different components of the pulmonary circulation. In this review, we explore advances in imaging of the pulmonary vasculature and their potential clinical translation. These include advances in diagnosis and assessing treatment response, as well as strategies that allow reduced radiation exposure and implementation of artificial intelligence technology. These emerging modalities hold the promise of developing a deeper understanding of pulmonary vascular disease and the impact of comorbidities. They also have the potential to improve patient outcomes by reducing time to diagnosis, refining classification, monitoring treatment response and improving our understanding of disease mechanisms.
Collapse
Affiliation(s)
| | - Harm J Bogaard
- Department of Pulmonology, Amsterdam University Medical Center, Location VU Medical Center, Amsterdam, The Netherlands
| | - Mohammed S M Elbaz
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin H Freed
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Edwin J R van Beek
- Edinburgh Imaging, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Deepa Gopalan
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - David G Kiely
- Sheffield Pulmonary Vascular Disease Unit and NIHR Biomedical Research Centre Sheffield, Royal Hallamshire Hospital, Sheffield, UK
| |
Collapse
|
3
|
Nappi AG, Santo G, Jonghi-Lavarini L, Miceli A, Lazzarato A, La Torre F, Dondi F, Gorica J. Emerging Role of [ 18F]FLT PET/CT in Lymphoid Malignancies: A Review of Clinical Results. Hematol Rep 2024; 16:32-41. [PMID: 38247994 PMCID: PMC10801569 DOI: 10.3390/hematolrep16010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Fluorine-18 fluorodeoxyglucose ([18F]FDG) is nowadays the leading positron emission tomography (PET) tracer for routine clinical work-ups in hematological malignancies; however, it is limited by false positive findings. Notably, false positives can occur in inflammatory and infective cases or in necrotic tumors that are infiltrated by macrophages and other inflammatory cells. In this context, 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) has been shown to be a promising imaging biomarker of hematological malignant cell proliferation. In this review, a total of 15 papers were reviewed to collect literature data regarding the clinical application of [18F]FLT PET/CT in hematological malignancies. This imaging modality seems to be a suitable tool for noninvasive assessment of tumor grading, also showing a correlation with Ki-67 immunostaining. Moreover, [18F]FLT PET/CT demonstrated high sensitivity in detecting aggressive lymphoma lesions, especially when applying a standardized uptake value (SUV) cutoff of 3. At baseline, the potential of [18F]FLT imaging as a predictive tool is demonstrated by the low tracer uptake in patients with a complete response. However, its use is limited in evaluating bone diseases due to its high physiological uptake in bone marrow. Interim [18F]FLT PET/CT (iFLT) has the potential to identify high-risk patients with greater precision than [18F]FDG PET/CT, optimizing risk-adapted therapy strategies. Moreover, [18F]FLT uptake showed a greater ability to differentiate tumor from inflammation compared to [18F]FDG, allowing the reduction of false-positive findings and making the first one a more selective tracer. Finally, FLT emerges as a superior independent predictor of PFS and OS compared to FDG and ensures a reliable early response assessment with greater accuracy and predictive value.
Collapse
Affiliation(s)
- Anna Giulia Nappi
- Section of Nuclear Medicine, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Giulia Santo
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzato, Italy;
| | | | - Alberto Miceli
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio E Biagio E Cesare Arrigo, 15121 Alessandria, Italy;
| | | | - Flavia La Torre
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Francesco Dondi
- Nuclear Medicine, ASST Spedali Civili Di Brescia and Università degli Studi di Brescia, 25123 Brescia, Italy
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy;
| |
Collapse
|
4
|
Liao S, Zhou M, Wang Y, Lu C, Yin B, Zhang Y, Liu H, Yin X, Song G. Emerging biomedical imaging-based companion diagnostics for precision medicine. iScience 2023; 26:107277. [PMID: 37520706 PMCID: PMC10371849 DOI: 10.1016/j.isci.2023.107277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
The tumor heterogeneity, which leads to individual variations in tumor microenvironments, causes poor prognoses and limits therapeutic response. Emerging technology such as companion diagnostics (CDx) detects biomarkers and monitors therapeutic responses, allowing identification of patients who would benefit most from treatment. However, currently, most US Food and Drug Administration-approved CDx tests are designed to detect biomarkers in vitro and ex vivo, making it difficult to dynamically report variations of targets in vivo. Various medical imaging techniques offer dynamic measurement of tumor heterogeneity and treatment response, complementing CDx tests. Imaging-based companion diagnostics allow for patient stratification for targeted medicines and identification of patient populations benefiting from alternative therapeutic methods. This review summarizes recent developments in molecular imaging for predicting and assessing responses to cancer therapies, as well as the various biomarkers used in imaging-based CDx tests. We hope this review provides informative insights into imaging-based companion diagnostics and advances precision medicine.
Collapse
Affiliation(s)
- Shiyi Liao
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Mengjie Zhou
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Youjuan Wang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Chang Lu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Baoli Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Ying Zhang
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Huiyi Liu
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Xia Yin
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| | - Guosheng Song
- State Key Laboratory for Chemo, Biosensing and Chemometrics, College of Chemistry and Chemical, Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
5
|
Abstract
MRI is a widely available clinical tool for cancer diagnosis and treatment monitoring. MRI provides excellent soft tissue imaging, using a wide range of contrast mechanisms, and can non-invasively detect tissue metabolites. These approaches can be used to distinguish cancer from normal tissues, to stratify tumor aggressiveness, and to identify changes within both the tumor and its microenvironment in response to therapy. In this review, the role of MRI in immunotherapy monitoring will be discussed and how it could be utilized in the future to address some of the unique clinical questions that arise from immunotherapy. For example, MRI could play a role in identifying pseudoprogression, mixed response, T cell infiltration, cell tracking, and some of the characteristic immune-related adverse events associated with these agents. The factors to be considered when developing MRI imaging biomarkers for immunotherapy will be reviewed. Finally, the advantages and limitations of each approach will be discussed, as well as the challenges for future clinical translation into routine clinical care. Given the increasing use of immunotherapy in a wide range of cancers and the ability of MRI to detect the microstructural and functional changes associated with successful response to immunotherapy, the technique has great potential for more widespread and routine use in the future for these applications.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Pippa G Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
6
|
Tatum JL, Kalen JD, Jacobs PM, Riffle LA, James A, Thang L, Sanders C, Hollingshead MG, Basuli F, Shi J, Doroshow JH. 3'-[ 18F]fluoro-3'-deoxythymidine ([ 18F]FLT) Positron Emission Tomography as an In Vivo Biomarker of inhibition of CDK 4/6-Rb pathway by Palbociclib in a patient derived bladder tumor. J Transl Med 2022; 20:375. [PMID: 35982453 PMCID: PMC9389794 DOI: 10.1186/s12967-022-03580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Several new generation CDK4/6 inhibitors have been developed and approved for breast cancer therapy in combination with endocrine therapeutics. Application of these inhibitors either alone or in combination in other solid tumors has been proposed, but no imaging biomarkers of response have been reported in non-breast cancer animal models. The purpose of this study was to evaluate 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT) Positron Emission Tomography (PET) as in vivo biomarker of response to palbociclib in a non-breast cancer model. Methods Twenty-four NSG mice bearing patient derived xenografts (PDX) of a well-characterized bladder tumor were randomized into 4 treatment groups: vehicle (n = 6); palbociclib (n = 6); temozolomide (n = 6); and palbociclib plus temozolomide (n = 6) and treated with two cycles of therapy or vehicle. Tumor uptake of [18F]FLT was determined by micro-PET/CT at baseline, 3 days, and 9 days post initiation of therapy. Following the second cycle of therapy, the mice were maintained until their tumors reached a size requiring humane termination. Results [18F]FLT uptake decreased significantly in the palbociclib and combination arms (p = 0.0423 and 0.0106 respectively at day 3 and 0.0012 and 0.0031 at day 9) with stable tumor volume. In the temozolomide arm [18F]FLT uptake increased with day 9 uptake significantly different than baseline (p = 0.0418) and progressive tumor growth was observed during the treatment phase. All groups exhibited progressive disease after day 22, 10 days following cessation of therapy. Conclusion Significant decreases in [18F]FLT uptake as early as three days post initiation of therapy with palbociclib, alone or in combination with temozolomide, in this bladder cancer model correlates with an absence of tumor growth during therapy that persists until day 18 for the palbociclib group and day 22 for the combination group (6 days and 10 days) following cessation of therapy. These results support early modulation of [18F]FLT as an in vivo biomarker predictive of palbociclib therapy response in a non-breast cancer model. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03580-8.
Collapse
Affiliation(s)
- James L Tatum
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Paula M Jacobs
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Lisa A Riffle
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Amy James
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Lai Thang
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Chelsea Sanders
- Animal Research Technical Support, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Melinda G Hollingshead
- Biological Testing Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, Frederick, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Edmonds CE, O'Brien SR, Mankoff DA, Pantel AR. Novel applications of molecular imaging to guide breast cancer therapy. Cancer Imaging 2022; 22:31. [PMID: 35729608 PMCID: PMC9210593 DOI: 10.1186/s40644-022-00468-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
The goals of precision oncology are to provide targeted drug therapy based on each individual’s specific tumor biology, and to enable the prediction and early assessment of treatment response to allow treatment modification when necessary. Thus, precision oncology aims to maximize treatment success while minimizing the side effects of inadequate or suboptimal therapies. Molecular imaging, through noninvasive assessment of clinically relevant tumor biomarkers across the entire disease burden, has the potential to revolutionize clinical oncology, including breast oncology. In this article, we review breast cancer positron emission tomography (PET) imaging biomarkers for providing early response assessment and predicting treatment outcomes. For 2-18fluoro-2-deoxy-D-glucose (FDG), a marker of cellular glucose metabolism that is well established for staging multiple types of malignancies including breast cancer, we highlight novel applications for early response assessment. We then review current and future applications of novel PET biomarkers for imaging the steroid receptors, including the estrogen and progesterone receptors, the HER2 receptor, cellular proliferation, and amino acid metabolism.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University if Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
8
|
Botros L, Jansen SM, Ashek A, Spruijt OA, Tramper J, Vonk Noordegraaf A, Aman J, Harms H, de Man FS, Huisman MC, Zhao L, Bogaard HJ. Application of [18F]FLT-PET in pulmonary arterial hypertension: a clinical study in pulmonary arterial hypertension patients and unaffected bone morphogenetic protein receptor type 2 mutation carriers. Pulm Circ 2021; 11:20458940211028017. [PMID: 34276963 PMCID: PMC8256252 DOI: 10.1177/20458940211028017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/05/2021] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension is a heterogeneous group of diseases characterized by vascular cell proliferation leading to pulmonary vascular remodelling and ultimately right heart failure. Previous data indicated that 3'-deoxy-3'-[18F]-fluorothymidine (18FLT) positron emission tomography (PET) scanning was increased in pulmonary arterial hypertension patients, hence providing a possible biomarker for pulmonary arterial hypertension as it reflects vascular cell hyperproliferation in the lung. This study sought to validate 18FLT-PET in an expanded cohort of pulmonary arterial hypertension patients in comparison to matched healthy controls and unaffected bone morphogenetic protein receptor type 2 mutation carriers. 18FLT-PET scanning was performed in 21 pulmonary arterial hypertension patients (15 hereditary pulmonary arterial hypertension and 6 idiopathic pulmonary arterial hypertension), 11 unaffected mutation carriers and 9 healthy control subjects. In-depth kinetic analysis indicated that there were no differences in lung 18FLT k3 phosphorylation among pulmonary arterial hypertension patients, unaffected bone morphogenetic protein receptor type 2 mutation carriers and healthy controls. Lung 18FLT uptake did not correlate with haemodynamic or clinical parameters in pulmonary arterial hypertension patients. Sequential 18FLT-PET scanning in three patients demonstrated uneven regional distribution in 18FLT uptake by 3D parametric mapping of the lung, although this did not follow the clinical course of the patient. We did not detect significantly increased lung 18FLT uptake in pulmonary arterial hypertension patients, nor in the unaffected bone morphogenetic protein receptor type 2 mutation carriers, as compared to healthy subjects. The conflicting results with our preliminary human 18FLT report may be explained by a small sample size previously and we observed large variation of lung 18FLT signals between patients, challenging the application of 18FLT-PET as a biomarker in the pulmonary arterial hypertension clinic.
Collapse
Affiliation(s)
- Liza Botros
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Samara M.A. Jansen
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Ali Ashek
- Faculty of MedicineNational Heart and Lung InstituteImperial College LondonHammersmith HospitalLondonUK
| | - Onno A. Spruijt
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Jelco Tramper
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Anton Vonk Noordegraaf
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Jurjan Aman
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Hans Harms
- Cardiovascular Imaging ProgramDepartments of Radiology and Medicine; Division of Nuclear Medicine and Molecular ImagingBrigham and Women’s Hospitaland Harvard Medical SchoolBostonMAUSA
- Institute of Clinical MedicineAarhus University HospitalAarhusDenmark
| | - Frances S. de Man
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Marc C. Huisman
- Department of Radiology and Nuclear MedicineAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Lan Zhao
- Faculty of MedicineNational Heart and Lung InstituteImperial College LondonHammersmith HospitalLondonUK
| | - Harm J. Bogaard
- Department of PulmonologyAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
9
|
Ventura M, Bernards N, De Souza R, Fricke IB, Hendriks BS, Fitzgerald JB, Lee H, Klinz SG, Zheng J. Longitudinal PET Imaging to Monitor Treatment Efficacy by Liposomal Irinotecan in Orthotopic Patient-Derived Pancreatic Tumor Models of High and Low Hypoxia. Mol Imaging Biol 2021; 22:653-664. [PMID: 31482415 PMCID: PMC7782415 DOI: 10.1007/s11307-019-01374-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Purpose Hypoxia is linked to aggressiveness, resistance to therapy, and poor prognosis of pancreatic tumors. Liposomal irinotecan (nal-IRI, ONIVYDE®) has shown potential in reducing hypoxia in the HT29 colorectal cancer model, and here, we investigate its therapeutic activity and ability to modulate hypoxia in patient-derived orthotopic tumor models of pancreatic cancer. Procedures Mice were randomized into nal-IRI treated and untreated controls. Magnetic resonance imaging was used for monitoring treatment efficacy, positron emission tomography (PET) imaging with F-18-labelled fluoroazomycinarabinoside ([18F]FAZA) for tumor hypoxia quantification, and F-18-labelled fluorothymidine ([18F]FLT) for tumor cell proliferation. Results The highly hypoxic OCIP51 tumors showed significant response following nal-IRI treatment compared with the less hypoxic OCIP19 tumors. [18F]FAZA-PET detected significant hypoxia reduction in treated OCIP51 tumors, 8 days before significant changes in tumor volume. OCIP19 tumors also responded to therapy, although tumor volume control was not accompanied by any reduction in [18F]FAZA uptake. In both models, no differences were observable in [18F]FLT uptake in treated tumors compared with control mice. Conclusions Hypoxia modulation may play a role in nal-IRI’s mechanism of action. Nal-IRI demonstrated greater anti-tumor activity in the more aggressive and hypoxic tumor model. Furthermore, hypoxia imaging provided early prediction of treatment response. Electronic supplementary material The online version of this article (10.1007/s11307-019-01374-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuela Ventura
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Nicholas Bernards
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Raquel De Souza
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Inga B Fricke
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | | | | | - Helen Lee
- Merrimack Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Stephan G Klinz
- Merrimack Pharmaceuticals, Inc., Cambridge, MA, USA
- Ipsen Bioscience, Cambridge, MA, USA
| | - Jinzi Zheng
- TECHNA Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada.
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
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
|
11
|
Gillman JA, Pantel AR, Mankoff DA, Edmonds CE. Update on Quantitative Imaging for Predicting and Assessing Response in Oncology. Semin Nucl Med 2020; 50:505-517. [PMID: 33059820 PMCID: PMC9788668 DOI: 10.1053/j.semnuclmed.2020.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging has revolutionized clinical oncology by imaging-specific facets of cancer biology. Through noninvasive measurements of tumor physiology, targeted radiotracers can serve as biomarkers for disease characterization, prognosis, response assessment, and predicting long-term response/survival. In turn, these imaging biomarkers can be utilized to tailor therapeutic regimens to tumor biology. In this article, we review biomarker applications for response assessment and predicting long-term outcomes. 18F-fluorodeoxyglucose (FDG), a measure of cellular glucose metabolism, is discussed in the context of lymphoma and breast and lung cancer. FDG has gained widespread clinical acceptance and has been integrated into the routine clinical care of several malignancies, most notably lymphoma. The novel radiotracers 16α-18F-fluoro-17β-estradiol and 18F-fluorothymidine are reviewed in application to the early prediction of response assessment of breast cancer. Through illustrative examples, we explore current and future applications of molecular imaging biomarkers in the advancement of precision medicine.
Collapse
Affiliation(s)
- Jennifer A Gillman
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - David A Mankoff
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Christine E Edmonds
- Department of Radiology, Division of Nuclear Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
12
|
Optimal Control Theory for Personalized Therapeutic Regimens in Oncology: Background, History, Challenges, and Opportunities. J Clin Med 2020; 9:jcm9051314. [PMID: 32370195 PMCID: PMC7290915 DOI: 10.3390/jcm9051314] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Optimal control theory is branch of mathematics that aims to optimize a solution to a dynamical system. While the concept of using optimal control theory to improve treatment regimens in oncology is not novel, many of the early applications of this mathematical technique were not designed to work with routinely available data or produce results that can eventually be translated to the clinical setting. The purpose of this review is to discuss clinically relevant considerations for formulating and solving optimal control problems for treating cancer patients. Our review focuses on two of the most widely used cancer treatments, radiation therapy and systemic therapy, as they naturally lend themselves to optimal control theory as a means to personalize therapeutic plans in a rigorous fashion. To provide context for optimal control theory to address either of these two modalities, we first discuss the major limitations and difficulties oncologists face when considering alternate regimens for their patients. We then provide a brief introduction to optimal control theory before formulating the optimal control problem in the context of radiation and systemic therapy. We also summarize examples from the literature that illustrate these concepts. Finally, we present both challenges and opportunities for dramatically improving patient outcomes via the integration of clinically relevant, patient-specific, mathematical models and optimal control theory.
Collapse
|
13
|
Carlin D, Weller A, Kramer G, Liu Y, Waterton JC, Chiti A, Sollini M, Joop de Langen A, O'Brien MER, Urbanowicz M, Jacobs BK, deSouza N. Evaluation of diffusion-weighted MRI and (18F) fluorothymidine-PET biomarkers for early response assessment in patients with operable non-small cell lung cancer treated with neoadjuvant chemotherapy. BJR Open 2019; 1:20190029. [PMID: 33178953 PMCID: PMC7592464 DOI: 10.1259/bjro.20190029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Objective: To correlate changes in the apparent diffusion coefficient (ADC) from diffusion-weighted (DW)-MRI and standardised uptake value (SUV) from fluorothymidine (18FLT)-PET/CT with histopathological estimates of response in patients with non-small cell lung cancer (NSCLC) treated with neoadjuvant chemotherapy and track longitudinal changes in these biomarkers in a multicentre, multivendor setting. Methods: 14 patients with operable NSCLC recruited to a prospective, multicentre imaging trial (EORTC-1217) were treated with platinum-based neoadjuvant chemotherapy. 13 patients had DW-MRI and FLT-PET/CT at baseline (10 had both), 12 were re-imaged at Day 14 (eight dual-modality) and nine after completing chemotherapy, immediately before surgery (six dual-modality). Surgical specimens (haematoxylin-eosin and Ki67 stained) estimated the percentage of residual viable tumour/necrosis and proliferation index. Results: Despite the small numbers,significant findings were possible. ADCmedian increased (p < 0.001) and SUVmean decreased (p < 0.001) significantly between baseline and Day 14; changes between Day 14 and surgery were less marked. All responding tumours (>30% reduction in unidimensional measurement pre-surgery), showed an increase at Day 14 in ADC75th centile and reduction in total lesion proliferation (SUVmean x proliferative volume) greater than established measurement variability. Change in imaging biomarkers did not correlate with histological response (residual viable tumour, necrosis). Conclusion: Changes in ADC and FLT-SUV following neoadjuvant chemotherapy in NSCLC were measurable by Day 14 and preceded changes in unidimensional size but did not correlate with histopathological response. However, the magnitude of the changes and their utility in predicting (non-) response (tumour size/clinical outcome) remains to be established. Advances in knowledge: During treatment, ADC increase precedes size reductions, but does not reflect histopathological necrosis.
Collapse
Affiliation(s)
- Dominic Carlin
- CRUK Imaging Centre, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | | | - Gem Kramer
- Department of Respiratory Diseases, VU University Medical Center, Amsterdam, The Netherlands
| | - Yan Liu
- EORTC Headquarters, Brussels, Belgium
| | - John C Waterton
- Centre for Imaging Sciences, Division of Informatics Imaging & Data Sciences, School of Health Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road Manchester M13 9PL UK
| | | | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Mary E R O'Brien
- The Royal Marsden Hospital, Downs Road, Sutton, Surrey SM2 5PT, UK
| | | | | | | |
Collapse
|
14
|
Evaluation of [ 18F]FDG/[ 18F]FLT/[ 18F]FMISO-based micro-positron emission tomography in detection of liver metastasis in human colorectal cancer. Nucl Med Biol 2019; 72-73:36-44. [PMID: 31330410 DOI: 10.1016/j.nucmedbio.2019.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/10/2019] [Accepted: 07/06/2019] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Positron emission tomography (PET) is extensively used in clinical oncology for tumor detection. This study aimed to explore the application of the radiotracers [18F]fluorodeoxyglucose ([18F]FDG), 3'-deoxy-3'- [18F]fluorothymidine ([18F]FLT), and [18F]fluoromisonidazole ([18F]FMISO) in the diagnosis and monitoring of hepatic metastasis in human colorectal cancer (CRC). METHODS A mouse model of human CRC with hepatic metastasis was established by intrasplenic implantation of human CRC cell lines LoVo or HCT8. Metastatic potential of these two cell lines was evaluated by wound healing assay in vitro and survival analysis. Uptake of radiotracers between LoVo and HCT8 cells and uptake of radiotracers in the resulting mouse tumor models were examined by in vivo and in vitro experiments. Uptake of each radiotracer in hepatic metastatic lesions was quantified and expressed as standard uptake value (SUV). Protein expression of multiple tumor biomarkers was determined in metastatic lesions. The correlation between tracer uptake and tumor marker expression was evaluated using linear regression. RESULTS LoVo cells exhibited a stronger metastatic potential and a higher radiotracer uptake ability than HCT8 cells, as evidenced by significantly greater wound closure percentage, shorter survival, higher incidence of liver metastases, and higher cellular radiotracer levels in LoVo cells or LoVo cell-xenografted mice. SUV values of [18F]FLT and [18F]FMISO, but not [18F]FDG, in LoVo cell-derived metastatic lesions were significantly greater than those in HCT8 lesions. Mechanistically, the expression of MACC1, HIF-1α, and GLUT-1(metastasis associated in colon cancer 1, MACC1; hypoxia-inducible factor 1-alpha, HIF-1α; and glucose transporter 1, GLUT-1, respectively) in LoVo cell-derived metastatic lesions was more effectively induced than in HCT8-derived ones. A linear regression analysis demonstrated significant positive correlations between [18F]FLT/[18F]FMISO uptake and tumor biomarker expression in metastatic tissues. CONCLUSIONS [18F]FLT and [18F]FMISO-based PET imaging may serve as a promising method for early detection and monitoring of hepatic metastasis in patients with CRC.
Collapse
|
15
|
Zanoni L, Broccoli A, Lambertini A, Pellegrini C, Stefoni V, Lodi F, Fonti C, Nanni C, Zinzani PL, Fanti S. Role of 18F-FLT PET/CT in suspected recurrent or residual lymphoma: final results of a pilot prospective trial. Eur J Nucl Med Mol Imaging 2019; 46:1661-1671. [DOI: 10.1007/s00259-019-04323-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/29/2019] [Indexed: 01/01/2023]
|
16
|
Elmi A, Makvandi M, Weng CC, Hou C, Clark AS, Mach RH, Mankoff DA. Cell-Proliferation Imaging for Monitoring Response to CDK4/6 Inhibition Combined with Endocrine-Therapy in Breast Cancer: Comparison of [ 18F]FLT and [ 18F]ISO-1 PET/CT. Clin Cancer Res 2019; 25:3063-3073. [PMID: 30692100 PMCID: PMC9788667 DOI: 10.1158/1078-0432.ccr-18-2769] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/11/2018] [Accepted: 01/14/2019] [Indexed: 12/25/2022]
Abstract
PURPOSE Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors in combination with endocrine-therapy have emerged as an important regimen of care for estrogen receptor (ER)-positive metastatic breast cancer, although identifying predictive biomarkers remains a challenge. We assessed the ability of two PET-proliferation tracers, [18F]FLT and [18F]ISO-1, for evaluating response to CDK4/6-inhibitor (palbociclib) and ER-antagonist (fulvestrant). EXPERIMENTAL DESIGN To determine the effect of CDK4/6 inhibition combined with estrogen-blockade, we assessed cell proliferation in six breast cancer cell lines after 1, 3, and 6 days of treatment with palbociclib and/or fulvestrant. These data were correlated to in vitro radiotracer assays and results were verified by longitudinal [18F]FLT and [18F]ISO-1 micro-PET imaging performed in MCF7 tumor-bearing mice. RESULTS All palbociclib-sensitive cell lines showed decreased [18F]FLT accumulation and S-phase depletion after treatment, with both measures augmented by combination therapy. In contrast, these cells showed changes in [18F]ISO-1 analogue-binding and G0 arrest only after prolonged treatment. MicroPET imaging of MCF7 xenografts showed a significant decrease in [18F]FLT but no changes in [18F]ISO-1 uptake in all treated mice on day 3. On day 14, however, mice treated with combination therapy showed a significant decrease in [18F]ISO-1, corresponding to G0 arrest, while maintaining reduced [18F]FLT uptake, which corresponded to S-phase depletion. CONCLUSIONS Our data suggest complementary roles of [18F]FLT and [18F]ISO-1 PET in evaluating tumor-proliferation after combined CDK4/6 inhibitor and endocrine therapy in breast cancer. [18F]FLT is more sensitive to immediate changes in S-phase, whereas [18F]ISO-1 can assess more delayed changes related to cell-cycle arrest and transition to G0 quiescence from combination therapy. These data suggest a potential role for early prediction of long-term response using these imaging biomarkers.
Collapse
Affiliation(s)
- Azadeh Elmi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehran Makvandi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chi-Chang Weng
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy S Clark
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
17
|
Frigerio B, Bizzoni C, Jansen G, Leamon CP, Peters GJ, Low PS, Matherly LH, Figini M. Folate receptors and transporters: biological role and diagnostic/therapeutic targets in cancer and other diseases. J Exp Clin Cancer Res 2019; 38:125. [PMID: 30867007 PMCID: PMC6417013 DOI: 10.1186/s13046-019-1123-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/28/2019] [Indexed: 01/28/2023] Open
Abstract
Folate receptors and transporters and one-carbon metabolism continue to be important areas of study given their essential roles in an assortment of diseases and as targets for treatment of cancer and inflammation. Reflecting this, every 2 years, the Folate Receptor Society organizes an international meeting, alternating between North America and Europe, where basic and translational scientists, clinical oncologists and rheumatologists from both academia and industry convene in an informal setting. The 7th International Symposium on Folate Receptors and Transporters was held in Sant'Alessio Siculo (ME), Taormina, Italy from 1st to 5th of October 2018, organized by Dr. Mariangela Figini from Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Following the format of previous meetings, more than 50 scientists from 9 different countries attended the 2018 meeting to share ongoing developments, discuss current research challenges and identify new avenues in basic and translational research. An important feature of this meeting was the participation of young investigators and trainees in this area, two (A. Dekhne and N. Verweij) of whom were awarded fellowships to attend this meeting as a recognition of the high scientific quality of their work. This report provides a synopsis of the highlights presented in the following sessions: Barton Kamen Lecture; Targeting one-carbon metabolism in cytosol and mitochondria; Structure and biology of the one-carbon solute transporters; Physiology and pathophysiology of folate receptors and transporters; Folate receptors for targeting tumors and inflammatory diseases; Conventional and new anti-folate drugs for treating inflammatory diseases and cancer; Imaging; Ongoing clinical trials; and Chimeric Antigen Receptor cell therapies of cancer.
Collapse
Affiliation(s)
- Barbara Frigerio
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Bizzoni
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Present address: ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, location Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Philip S. Low
- Purdue University Institute for Drug Discovery, West Lafayette, Indiana, USA
| | - Larry H. Matherly
- Barbara Ann Karmanos Cancer Institute and Wayne State University School of Medicine, Detroit, MI USA
| | - Mariangela Figini
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
18
|
Jarrett AM, Lima EABF, Hormuth DA, McKenna MT, Feng X, Ekrut DA, Resende ACM, Brock A, Yankeelov TE. Mathematical models of tumor cell proliferation: A review of the literature. Expert Rev Anticancer Ther 2018; 18:1271-1286. [PMID: 30252552 PMCID: PMC6295418 DOI: 10.1080/14737140.2018.1527689] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION A defining hallmark of cancer is aberrant cell proliferation. Efforts to understand the generative properties of cancer cells span all biological scales: from genetic deviations and alterations of metabolic pathways to physical stresses due to overcrowding, as well as the effects of therapeutics and the immune system. While these factors have long been studied in the laboratory, mathematical and computational techniques are being increasingly applied to help understand and forecast tumor growth and treatment response. Advantages of mathematical modeling of proliferation include the ability to simulate and predict the spatiotemporal development of tumors across multiple experimental scales. Central to proliferation modeling is the incorporation of available biological data and validation with experimental data. Areas covered: We present an overview of past and current mathematical strategies directed at understanding tumor cell proliferation. We identify areas for mathematical development as motivated by available experimental and clinical evidence, with a particular emphasis on emerging, non-invasive imaging technologies. Expert commentary: The data required to legitimize mathematical models are often difficult or (currently) impossible to obtain. We suggest areas for further investigation to establish mathematical models that more effectively utilize available data to make informed predictions on tumor cell proliferation.
Collapse
Affiliation(s)
- Angela M Jarrett
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
- b Livestrong Cancer Institutes , The University of Texas at Austin , Austin , USA
| | - Ernesto A B F Lima
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
| | - David A Hormuth
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
- b Livestrong Cancer Institutes , The University of Texas at Austin , Austin , USA
| | - Matthew T McKenna
- c Department of Biomedical Engineering , Vanderbilt University , Nashville , USA
| | - Xinzeng Feng
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
| | - David A Ekrut
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
| | - Anna Claudia M Resende
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
- d Department of Computational Modeling , National Laboratory for Scientific Computing , Petrópolis , Brazil
| | - Amy Brock
- b Livestrong Cancer Institutes , The University of Texas at Austin , Austin , USA
- e Department of Biomedical Engineering , The University of Texas at Austin , Austin , USA
| | - Thomas E Yankeelov
- a Institute for Computational Engineering and Sciences , The University of Texas at Austin , Austin , USA
- b Livestrong Cancer Institutes , The University of Texas at Austin , Austin , USA
- e Department of Biomedical Engineering , The University of Texas at Austin , Austin , USA
- f Department of Diagnostic Medicine , The University of Texas at Austin , Austin , USA
- g Department of Oncology , The University of Texas at Austin , Austin , USA
| |
Collapse
|
19
|
Gock M, Mullins CS, Bergner C, Prall F, Ramer R, Göder A, Krämer OH, Lange F, Krause BJ, Klar E, Linnebacher M. Establishment, functional and genetic characterization of three novel patient-derived rectal cancer cell lines. World J Gastroenterol 2018; 24:4880-4892. [PMID: 30487698 PMCID: PMC6250916 DOI: 10.3748/wjg.v24.i43.4880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To establish patient-individual tumor models of rectal cancer for analyses of novel biomarkers, individual response prediction and individual therapy regimens. METHODS Establishment of cell lines was conducted by direct in vitro culturing and in vivo xenografting with subsequent in vitro culturing. Cell lines were in-depth characterized concerning morphological features, invasive and migratory behavior, phenotype, molecular profile including mutational analysis, protein expression, and confirmation of origin by DNA fingerprint. Assessment of chemosensitivity towards an extensive range of current chemotherapeutic drugs and of radiosensitivity was performed including analysis of a combined radio- and chemotherapeutic treatment. In addition, glucose metabolism was assessed with 18F-fluorodeoxyglucose (FDG) and proliferation with 18F-fluorothymidine. RESULTS We describe the establishment of ultra-low passage rectal cancer cell lines of three patients suffering from rectal cancer. Two cell lines (HROC126, HROC284Met) were established directly from tumor specimens while HROC239 T0 M1 was established subsequent to xenografting of the tumor. Molecular analysis classified all three cell lines as CIMP-0/ non-MSI-H (sporadic standard) type. Mutational analysis revealed following mutational profiles: HROC126: APCwt , TP53wt , KRASwt , BRAFwt , PTENwt ; HROC239 T0 M1: APCmut , P53wt , KRASmut , BRAFwt , PTENmut and HROC284Met: APCwt , P53mut , KRASmut , BRAFwt , PTENmut . All cell lines could be characterized as epithelial (EpCAM+) tumor cells with equivalent morphologic features and comparable growth kinetics. The cell lines displayed a heterogeneous response toward chemotherapy, radiotherapy and their combined application. HROC126 showed a highly radio-resistant phenotype and HROC284Met was more susceptible to a combined radiochemotherapy than HROC126 and HROC239 T0 M1. Analysis of 18F-FDG uptake displayed a markedly reduced FDG uptake of all three cell lines after combined radiochemotherapy. CONCLUSION These newly established and in-depth characterized ultra-low passage rectal cancer cell lines provide a useful instrument for analysis of biological characteristics of rectal cancer.
Collapse
Affiliation(s)
- Michael Gock
- Department of General Surgery, University Medical Center, Rostock 18055, Germany
| | - Christina S Mullins
- Section of Molecular Oncology and Immunotherapy, University Medical Center, Rostock 18055, Germany
| | - Carina Bergner
- Department of Nuclear Medicine, University Medical Center, Rostock 18055, Germany
| | - Friedrich Prall
- Institute of Pathology, University Medical Center, Rostock 18055, Germany
| | - Robert Ramer
- Institute of Pharmacology, University Medical Center, Rostock 18055, Germany
| | - Anja Göder
- Institute of Toxicology, University Medical Center Mainz, Mainz 55131, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center Mainz, Mainz 55131, Germany
| | - Falko Lange
- Oscar-Langendorff-Institute of Physiology, University Medical Center, Rostock 18055, Germany
| | - Bernd J Krause
- Department of Nuclear Medicine, University Medical Center, Rostock 18055, Germany
| | - Ernst Klar
- Department of General Surgery, University Medical Center, Rostock 18055, Germany
| | - Michael Linnebacher
- Section of Molecular Oncology and Immunotherapy, University Medical Center, Rostock 18055, Germany
| |
Collapse
|
20
|
Duan X, Zhang X, Gan Q, Fang S, Ruan Q, Song X, Zhang J. Novel 99mTc-labelled complexes with thymidine isocyanide: radiosynthesis and evaluation as potential tumor imaging tracers. MEDCHEMCOMM 2018; 9:705-712. [PMID: 30108961 PMCID: PMC6071732 DOI: 10.1039/c7md00635g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/27/2018] [Indexed: 01/11/2023]
Abstract
A novel thymidine isocyanide (CN-TdR) functionalized at the N3 position of thymidine was synthesized and then radiolabelled with 99mTc(i) and [99mTc(i)(CO)3]+ cores to produce [99mTc(CN-TdR)6]+ and [99mTc(CO)3(CN-TdR)3]+, respectively. Both of them were prepared with high radiochemical purity and were stable over 6 h in saline at ambient temperature and in serum at 37 °C. The partition coefficient results demonstrated that they were hydrophilic. The cell internalization studies showed that their uptake might be mediated by nucleoside transporters. Biodistribution of these complexes in mice bearing the S180 tumor showed that they accumulated in the tumor with high uptake and cleared rapidly from blood and muscles, producing high tumor/blood and tumor/muscle ratios. Between them, [99mTc(CN-TdR)6]+ exhibited advantages concerning a higher tumor uptake, tumor/blood ratio and tumor/muscle ratio at 60 min post-injection. Single photon emission computed tomography imaging studies showed that there was a clear accumulation in tumor sites, suggesting that [99mTc(CN-TdR)6]+ could be a promising candidate for tumor imaging.
Collapse
Affiliation(s)
- Xiaojiang Duan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Xuran Zhang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
- Department of Isotopes , China Institute of Atomic Energy , P. O. Box 2108 , Beijing 102413 , P.R. China
| | - Qianqian Gan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Si'an Fang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Xiaoqing Song
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals , Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , P.R. China .
| |
Collapse
|
21
|
Xu HL, Li M, Zhang RJ, Jiang HJ, Zhang MY, Li X, Wang YQ, Pan WB. Prediction of tumor biological characteristics in different colorectal cancer liver metastasis animal models using 18F-FDG and 18F-FLT. Hepatobiliary Pancreat Dis Int 2018; 17:140-148. [PMID: 29571649 DOI: 10.1016/j.hbpd.2018.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Positron emission tomography (PET) is a noninvasive method to characterize different metabolic activities of tumors, providing information for staging, prognosis, and therapeutic response of patients with cancer. The aim of this study was to evaluate the feasibility of 18F-fludeoxyglucose (18F-FDG) and 3'-deoxy-3'-18F-fluorothymidine (18F-FLT) PET in predicting tumor biological characteristics of colorectal cancer liver metastasis. METHODS The uptake rate of 18F-FDG and 18F-FLT in SW480 and SW620 cells was measured via an in vitro cell uptake assay. The region of interest was drawn over the tumor and liver to calculate the maximum standardized uptake value ratio (tumor/liver) from PET images in liver metastasis model. The correlation between tracer uptake in liver metastases and VEGF, Ki67 and CD44 expression was evaluated by linear regression. RESULTS Compared to SW620 tumor-bearing mice, SW480 tumor-bearing mice presented a higher rate of liver metastases. The uptake rate of 18F-FDG in SW480 and SW620 cells was 6.07% ± 1.19% and 2.82% ± 0.15%, respectively (t = 4.69, P = 0.04); that of 18F-FLT was 24.81% ± 0.45% and 15.57% ± 0.66%, respectively (t = 19.99, P < 0.001). Micro-PET scan showed that all parameters of FLT were significantly higher in SW480 tumors than those in SW620 tumors. A moderate relationship was detected between metastases in the liver and 18F-FLT uptake in primary tumors (r = 0.73, P = 0.0019). 18F-FLT uptake was also positively correlated with the expression of CD44 in liver metastases (r = 0.81, P = 0.0049). CONCLUSIONS The uptake of 18F-FLT in metastatic tumor reflects different biological behaviors of colon cancer cells. 18F-FLT can be used to evaluate the metastatic potential of colorectal cancer in nude mice.
Collapse
Affiliation(s)
- Hai-Long Xu
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Man Li
- Endoscopy Center, the Third Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Rong-Jun Zhang
- Key Laboratory of Nuclear Medicine of the Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Wuxi 214063, China
| | - Hui-Jie Jiang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China.
| | - Ming-Yu Zhang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Li
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yi-Qiao Wang
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wen-Bin Pan
- Department of Radiology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
22
|
Rapic S, Vangestel C, Verhaeghe J, Thomae D, Pauwels P, Van den Wyngaert T, Staelens S, Stroobants S. Evaluation of [ 18F]Fluorothymidine as a Biomarker for Early Therapy Response in a Mouse Model of Colorectal Cancer. Mol Imaging Biol 2017; 19:109-119. [PMID: 27324368 DOI: 10.1007/s11307-016-0974-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE In oncology, positron emission tomography imaging using dedicated tracers as biomarkers may assist in early evaluation of therapy efficacy. Using 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT), we investigated the early effects of chemotherapeutic treatment on cancer cell proliferation in a BRAF-mutated colorectal cancer xenograft model. PROCEDURES Colo205 subcutaneously inoculated animals underwent 90-min dynamic imaging before and 24 h after treatment with vehicle (control), cetuximab (resistant) or irinotecan (sensitive). Total distribution volume was quantified from dynamic data, and standardized uptake values as well as tumor-to-blood ratios were calculated from static images averaged over the last 20 min. In vivo imaging data was correlated with ex vivo proliferation and thymidine metabolism proteins. RESULTS All imaging parameters showed a significant post-treatment decrease from [18F]FLT baseline uptake for the irinotecan group (p ≤ 0.001) as compared with the cetuximab and vehicle group and correlated strongly with each other (p ≤ 0.0001). In vivo data were in agreement with Ki67 staining, showing a significantly lower percentage of Ki67-positive cells in the irinotecan group as compared with other groups (p ≤ 0.0001). Tumor expression of thymidine kinase 1 phosphorylated on serine 13, thymidylate synthase, and thymidine phosphorylase remained unaffected, while thymidine kinase 1 expression was, surprisingly, significantly higher in irinotecan-treated animals (p ≤ 0.01). In contrast, tumor ATP levels were lowest in this group. CONCLUSIONS [18F]FLT positron emission tomography was found to be a suitable biomarker of early tumor response to anti-proliferative treatment, with static imaging not being inferior to full compartmental analysis in our xenograft model. The dynamics of thymidine kinase 1 protein expression and protein activity in low ATP environments merits further investigation.
Collapse
Affiliation(s)
- Sara Rapic
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - David Thomae
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Patrick Pauwels
- Center for Oncological Research (CORE), University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Tim Van den Wyngaert
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
- Department of Nuclear Medicine, Antwerp University Hospital, Wilrijkstraat 10, 2650, Edegem, Belgium.
| |
Collapse
|
23
|
Heskamp S, Heijmen L, Gerrits D, Molkenboer-Kuenen JDM, Ter Voert EGW, Heinzmann K, Honess DJ, Smith DM, Griffiths JR, Doblas S, Sinkus R, Laverman P, Oyen WJG, Heerschap A, Boerman OC. Response Monitoring with [ 18F]FLT PET and Diffusion-Weighted MRI After Cytotoxic 5-FU Treatment in an Experimental Rat Model for Colorectal Liver Metastases. Mol Imaging Biol 2017; 19:540-549. [PMID: 27798786 PMCID: PMC5498638 DOI: 10.1007/s11307-016-1021-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE The aim of the study was to investigate the potential of diffusion-weighted magnetic resonance imaging (DW-MRI) and 3'-dexoy-3'-[18F]fluorothymidine ([18F]FLT) positron emission tomography (PET) as early biomarkers of treatment response of 5-fluorouracil (5-FU) in a syngeneic rat model of colorectal cancer liver metastases. PROCEDURES Wag/Rij rats with intrahepatic syngeneic CC531 tumors were treated with 5-FU (15, 30, or 60 mg/kg in weekly intervals). Before treatment and at days 1, 3, 7, and 14 after treatment rats underwent DW-MRI and [18F]FLT PET. Tumors were analyzed immunohistochemically for Ki67, TK1, and ENT1 expression. RESULTS 5-FU inhibited the growth of CC531 tumors in a dose-dependent manner. Immunohistochemical analysis did not show significant changes in Ki67, TK1, and ENT1 expression. However, [18F]FLT SUVmean and SUVmax were significantly increased at days 4 and 7 after treatment with 5-FU (60 mg/kg) and returned to baseline at day 14 (SUVmax at days -1, 4, 7, and 14 was 1.1 ± 0.1, 2.3 ± 0.5, 2.3 ± 0.6, and 1.5 ± 0.4, respectively). No changes in [18F]FLT uptake were observed in the nontreated animals. Furthermore, the apparent diffusion coefficient (ADCmean) did not change in 5-FU-treated rats compared to untreated rats. CONCLUSION This study suggests that 5-FU treatment induces a flare in [18F]FLT uptake of responsive CC531 tumors in the liver, while the ADCmean did not change significantly. Future studies in larger groups are warranted to further investigate whether [18F]FLT PET can discriminate between disease progression and treatment response.
Collapse
Affiliation(s)
- Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Linda Heijmen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danny Gerrits
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Edwin G W Ter Voert
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kathrin Heinzmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Davina J Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Sabrina Doblas
- LBI, CRI - UMR 1149 Inserm, Université Paris Diderot, Paris, France
| | - Ralph Sinkus
- BHF Centre of Excellence, Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, UK
| | - Peter Laverman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Schelhaas S, Heinzmann K, Bollineni VR, Kramer GM, Liu Y, Waterton JC, Aboagye EO, Shields AF, Soloviev D, Jacobs AH. Preclinical Applications of 3'-Deoxy-3'-[ 18F]Fluorothymidine in Oncology - A Systematic Review. Theranostics 2017; 7:40-50. [PMID: 28042315 PMCID: PMC5196884 DOI: 10.7150/thno.16676] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/16/2016] [Indexed: 11/05/2022] Open
Abstract
The positron emission tomography (PET) tracer 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) has been proposed to measure cell proliferation non-invasively in vivo. Hence, it should provide valuable information for response assessment to tumor therapies. To date, [18F]FLT uptake has found limited use as a response biomarker in clinical trials in part because a better understanding is needed of the determinants of [18F]FLT uptake and therapy-induced changes of its retention in the tumor. In this systematic review of preclinical [18F]FLT studies, comprising 174 reports, we identify the factors governing [18F]FLT uptake in tumors, among which thymidine kinase 1 plays a primary role. The majority of publications (83 %) report that decreased [18F]FLT uptake reflects the effects of anticancer therapies. 144 times [18F]FLT uptake was related to changes in proliferation as determined by ex vivo analyses. Of these approaches, 77 % describe a positive relation, implying a good concordance of tracer accumulation and tumor biology. These preclinical data indicate that [18F]FLT uptake holds promise as an imaging biomarker for response assessment in clinical studies. Understanding of the parameters which influence cellular [18F]FLT uptake and retention as well as the mechanism of changes induced by therapy is essential for successful implementation of this PET tracer. Hence, our systematic review provides the background for the use of [18F]FLT in future clinical studies.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | | | - Vikram R Bollineni
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Yan Liu
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, UK
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Dmitry Soloviev
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany.; Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany
| |
Collapse
|
25
|
Schelhaas S, Held A, Wachsmuth L, Hermann S, Honess DJ, Heinzmann K, Smith DM, Griffiths JR, Faber C, Jacobs AH. Gemcitabine Mechanism of Action Confounds Early Assessment of Treatment Response by 3'-Deoxy-3'-[18F]Fluorothymidine in Preclinical Models of Lung Cancer. Cancer Res 2016; 76:7096-7105. [PMID: 27784748 DOI: 10.1158/0008-5472.can-16-1479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/13/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
3'-Deoxy-3'-[18F]fluorothymidine positron emission tomography ([18F]FLT-PET) and diffusion-weighted MRI (DW-MRI) are promising approaches to monitor tumor therapy response. Here, we employed these two imaging modalities to evaluate the response of lung carcinoma xenografts in mice after gemcitabine therapy. Caliper measurements revealed that H1975 xenografts responded to gemcitabine treatment, whereas A549 growth was not affected. In both tumor models, uptake of [18F]FLT was significantly reduced 6 hours after drug administration. On the basis of the gemcitabine concentration and [18F]FLT excretion measured, this was presumably related to a direct competition of gemcitabine with the radiotracer for cellular uptake. On day 1 after therapy, [18F]FLT uptake was increased in both models, which was correlated with thymidine kinase 1 (TK1) expression. Two and 3 days after drug administration, [18F]FLT uptake as well as TK1 and Ki67 expression were unchanged. A reduction in [18F]FLT in the responsive H1975 xenografts could only be noted on day 5 of therapy. Changes in ADCmean in A549 xenografts 1 or 2 days after gemcitabine did not seem to be of therapy-related biological relevance as they were not related to cell death (assessed by caspase-3 IHC and cellular density) or tumor therapy response. Taken together, in these models, early changes of [18F]FLT uptake in tumors reflected mechanisms, such as competing gemcitabine uptake or gemcitabine-induced thymidylate synthase inhibition, and only reflected growth-inhibitory effects at a later time point. Hence, the time point for [18F]FLT-PET imaging of tumor response to gemcitabine is of crucial importance. Cancer Res; 76(24); 7096-105. ©2016 AACR.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Annelena Held
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Lydia Wachsmuth
- Department of Clinical Radiology, University Hospital of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Davina J Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kathrin Heinzmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Donna-Michelle Smith
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - John R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital of Münster, Münster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany.
- Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany
| |
Collapse
|
26
|
Kwee TC, Gholami S, Werner TJ, Rubello D, Alavi A, Høilund-Carlsen PF. 18F-FDG, as a single imaging agent in assessing cancer, shows the ongoing biological phenomena in many domains: do we need additional tracers for clinical purposes? Nucl Med Commun 2016; 37:333-7. [PMID: 26796033 DOI: 10.1097/mnm.0000000000000478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Thomas C Kwee
- aDepartment of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands bDepartment of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA cDepartment of Nuclear Medicine, PET/CT Centre, Radiology, Interventional Radiology NeuroRadiology, Medical Physics, Clinical Laboratory, Biomarkers Laboratory, Pathology, Microbiology, 'Santa Maria della Misericordia' Hospital, Rovigo, Italy dDepartment of Nuclear Medicine, Odense University Hospital eInstitute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | | |
Collapse
|
27
|
Heinzmann K, Honess DJ, Lewis DY, Smith DM, Cawthorne C, Keen H, Heskamp S, Schelhaas S, Witney TH, Soloviev D, Williams KJ, Jacobs AH, Aboagye EO, Griffiths JR, Brindle KM. The relationship between endogenous thymidine concentrations and [(18)F]FLT uptake in a range of preclinical tumour models. EJNMMI Res 2016; 6:63. [PMID: 27515446 PMCID: PMC4980847 DOI: 10.1186/s13550-016-0218-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Recent studies have shown that 3'-deoxy-3'-[(18)F] fluorothymidine ([(18)F]FLT)) uptake depends on endogenous tumour thymidine concentration. The purpose of this study was to investigate tumour thymidine concentrations and whether they correlated with [(18)F]FLT uptake across a broad spectrum of murine cancer models. A modified liquid chromatography-mass spectrometry (LC-MS/MS) method was used to determine endogenous thymidine concentrations in plasma and tissues of tumour-bearing and non-tumour bearing mice and rats. Thymidine concentrations were determined in 22 tumour models, including xenografts, syngeneic and spontaneous tumours, from six research centres, and a subset was compared for [(18)F]FLT uptake, described by the maximum and mean tumour-to-liver uptake ratio (TTL) and SUV. RESULTS The LC-MS/MS method used to measure thymidine in plasma and tissue was modified to improve sensitivity and reproducibility. Thymidine concentrations determined in the plasma of 7 murine strains and one rat strain were between 0.61 ± 0.12 μM and 2.04 ± 0.64 μM, while the concentrations in 22 tumour models ranged from 0.54 ± 0.17 μM to 20.65 ± 3.65 μM. TTL at 60 min after [(18)F]FLT injection, determined in 14 of the 22 tumour models, ranged from 1.07 ± 0.16 to 5.22 ± 0.83 for the maximum and 0.67 ± 0.17 to 2.10 ± 0.18 for the mean uptake. TTL did not correlate with tumour thymidine concentrations. CONCLUSIONS Endogenous tumour thymidine concentrations alone are not predictive of [(18)F]FLT uptake in murine cancer models.
Collapse
Affiliation(s)
- Kathrin Heinzmann
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Present address: Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
| | - Davina Jean Honess
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - David Yestin Lewis
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
| | | | - Christopher Cawthorne
- Wolfson Molecular Imaging Centre, Manchester Pharmacy School, University of Manchester, Manchester, UK
- Present address: Positron Emission Tomography Research Centre, University of Hull, Hull, UK
| | - Heather Keen
- Personalised Healthcare and Biomarkers, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Sandra Heskamp
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU), University Hospital of Münster, Münster, Germany
| | - Timothy Howard Witney
- Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
- Present address: UCL Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Dmitry Soloviev
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
| | - Kaye Janine Williams
- Wolfson Molecular Imaging Centre, Manchester Pharmacy School, University of Manchester, Manchester, UK
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK
| | - Andreas Hans Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU), University Hospital of Münster, Münster, Germany
| | - Eric Ofori Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, London, UK
| | | | - Kevin Michael Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- CRUK-EPSRC Cancer Imaging Centre in Cambridge and Manchester, Cambridge, UK.
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
28
|
McHugh CI, Lawhorn-Crews JM, Modi D, Douglas KA, Jones SK, Mangner TJ, Collins JM, Shields AF. Effects of capecitabine treatment on the uptake of thymidine analogs using exploratory PET imaging agents: 18F-FAU, 18F-FMAU, and 18F-FLT. Cancer Imaging 2016; 16:34. [PMID: 27751167 PMCID: PMC5067904 DOI: 10.1186/s40644-016-0092-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/03/2016] [Indexed: 12/02/2022] Open
Abstract
Background A principal goal for the use of positron emission tomography (PET) in oncology is for real-time evaluation of tumor response to chemotherapy. Given that many contemporary anti-neoplastic agents function by impairing cellular proliferation, it is of interest to develop imaging modalities to monitor these pathways. Here we examined the effect of capecitabine on the uptake of thymidine analogs used with PET: 3’-deoxy-3’-[18F]fluorothymidine (18F-FLT), 1-(2’-deoxy-2’-[18F]fluoro-β-D-arabinofuranosyl) thymidine (18F-FMAU), and 1-(2’-deoxy-2’-[18F]fluoro-β-D-arabinofuranosyl) uracil (18F-FAU) in patients with advanced cancer. Methods Fifteen patients were imaged, five with each imaging agent. Patients had been previously diagnosed with breast, colorectal, gastric, and esophageal cancers and had not received therapy for at least 4 weeks prior to the first scan, and had not been treated with any prior fluoropyrimidines. Subjects were imaged within a week before the start of capecitabine and on the second day of treatment, after the third dose of capecitabine. Tracer uptake was quantified by mean standard uptake value (SUVmean) and using kinetic analysis. Results Patients imaged with 18F-FLT showed variable changes in retention and two patients exhibited an increase in SUVmean of 172.3 and 89.9 %, while the other patients had changes ranging from +19.4 to -25.4 %. The average change in 18F-FMAU retention was 0.2 % (range -24.4 to 23.1) and 18F-FAU was -10.2 % (range -40.3 to 19.2). Observed changes correlated strongly with SUVmax but not kinetic measurements. Conclusions This pilot study demonstrates that patients treated with capecitabine can produce a marked increase in 18F-FLT retention in some patients, which will require further study to determine if this flare is predictive of therapeutic response. 18F-FAU and 18F-FMAU showed little change, on average, after treatment. Electronic supplementary material The online version of this article (doi:10.1186/s40644-016-0092-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher I McHugh
- Cancer Biology Graduate Program, Wayne State University, Detroit, MI, 48201, USA
| | - Jawana M Lawhorn-Crews
- Karmanos Cancer Institute and Oncology, Wayne State University, 4100 John R., HW04HO, Detroit, MI, 48201, USA
| | - Dipenkumar Modi
- Karmanos Cancer Institute and Oncology, Wayne State University, 4100 John R., HW04HO, Detroit, MI, 48201, USA
| | - Kirk A Douglas
- Karmanos Cancer Institute and Oncology, Wayne State University, 4100 John R., HW04HO, Detroit, MI, 48201, USA
| | - Steven K Jones
- Cancer Biology Graduate Program, Wayne State University, Detroit, MI, 48201, USA
| | | | | | - Anthony F Shields
- Karmanos Cancer Institute and Oncology, Wayne State University, 4100 John R., HW04HO, Detroit, MI, 48201, USA.
| |
Collapse
|
29
|
Martelli C, Dico AL, Diceglie C, Lucignani G, Ottobrini L. Optical imaging probes in oncology. Oncotarget 2016; 7:48753-48787. [PMID: 27145373 PMCID: PMC5217050 DOI: 10.18632/oncotarget.9066] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/10/2016] [Indexed: 01/19/2023] Open
Abstract
Cancer is a complex disease, characterized by alteration of different physiological molecular processes and cellular features. Keeping this in mind, the possibility of early identification and detection of specific tumor biomarkers by non-invasive approaches could improve early diagnosis and patient management.Different molecular imaging procedures provide powerful tools for detection and non-invasive characterization of oncological lesions. Clinical studies are mainly based on the use of computed tomography, nuclear-based imaging techniques and magnetic resonance imaging. Preclinical imaging in small animal models entails the use of dedicated instruments, and beyond the already cited imaging techniques, it includes also optical imaging studies. Optical imaging strategies are based on the use of luminescent or fluorescent reporter genes or injectable fluorescent or luminescent probes that provide the possibility to study tumor features even by means of fluorescence and luminescence imaging. Currently, most of these probes are used only in animal models, but the possibility of applying some of them also in the clinics is under evaluation.The importance of tumor imaging, the ease of use of optical imaging instruments, the commercial availability of a wide range of probes as well as the continuous description of newly developed probes, demonstrate the significance of these applications. The aim of this review is providing a complete description of the possible optical imaging procedures available for the non-invasive assessment of tumor features in oncological murine models. In particular, the characteristics of both commercially available and newly developed probes will be outlined and discussed.
Collapse
Affiliation(s)
- Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Umberto Veronesi Foundation, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Tecnomed Foundation, University of Milan-Bicocca, Monza, Italy
| | - Giovanni Lucignani
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Institute for Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| |
Collapse
|
30
|
Lai YL, Wu CY, Chao KSC. Biological imaging in clinical oncology: radiation therapy based on functional imaging. Int J Clin Oncol 2016; 21:626-632. [PMID: 27384183 DOI: 10.1007/s10147-016-1000-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/29/2016] [Indexed: 12/25/2022]
Abstract
Radiation therapy is one of the most effective tools for cancer treatment. In recent years, intensity-modulated radiation therapy has become increasingly popular in that target dose-escalation can be done while sparing adjacent normal tissues. For this reason, the development of measures to pave the way for accurate target delineation is of great interest. With the integration of functional information obtained by biological imaging with radiotherapy, strategies using advanced biological imaging to visualize metabolic pathways and to improve therapeutic index and predict treatment response are discussed in this article.
Collapse
Affiliation(s)
- Yo-Liang Lai
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - K S Clifford Chao
- China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| |
Collapse
|
31
|
Capistrano R, Vangestel C, Wouters A, Dockx Y, Pauwels P, Stroobants S, Apers S, Lardon F, Pieters L, Staelens S. Efficacy Screening of Gloriosa Superba Extracts in a Murine Pancreatic Cancer Model Using (18)F-FDG PET/CT for Monitoring Treatment Response. Cancer Biother Radiopharm 2016; 31:99-109. [PMID: 27093343 DOI: 10.1089/cbr.2015.1954] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE In vivo efficacy of two herbal extracts of Gloriosa superba L. (Colchicaceae) was investigated in a murine pancreatic tumor model by tumor volume measurements and Positron Emission Tomography (PET) imaging using 2-deoxy-2-[(18)F]fluoro-d-glucose ((18)F-FDG). MATERIALS AND METHODS A crude extract of G. superba (GS) seeds rich in colchicine and a colchicine-poor extract (GS2B) containing mostly colchicoside as a putative prodrug were prepared. PANC02-bearing C57BL/6 mice were treated with either placebo, gemcitabine, or one of the extracts (three different doses) for 10 days. Tumor volume measurements were performed daily during treatment and additionally (18)F-FDG Positron emission tomography/computed tomography was acquired at baseline and after 7 days of treatment. Ki-67 and cleaved caspase-3 immunostaining was performed on the resected tumors. RESULTS After 7 days of treatment, a dose-dependent tumor growth inhibition of both extracts was observed with the highest in vivo response at the highest dose of GS and GS2B and gemcitabine. A positive significant correlation was found between Ki-67 scores and relative tumor volumes (RTV), and a negative significant correlation between caspase-3 staining scores and RTV. A decrease in (18)F-FDG uptake was clearly observed in all treatment groups. CONCLUSIONS The therapeutic efficacy of the two different herbal extracts was demonstrated in an in vivo pancreatic tumor model. (18)F-FDG PET was able to detect an early response as overall lower (18)F-FDG uptake was measured in the treated groups.
Collapse
Affiliation(s)
- Rica Capistrano
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Christel Vangestel
- 2 Department of Nuclear Medicine, Antwerp University Hospital , Edegem, Belgium .,3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - An Wouters
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Yanina Dockx
- 3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - Patrick Pauwels
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Sigrid Stroobants
- 2 Department of Nuclear Medicine, Antwerp University Hospital , Edegem, Belgium .,3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| | - Sandra Apers
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Filip Lardon
- 4 Center for Oncological Research (CORE), University of Antwerp , Antwerp, Belgium
| | - Luc Pieters
- 1 Department of Pharmaceutical Sciences, Natural Products & Food Research and Analysis (NatuRA), University of Antwerp , Antwerp, Belgium
| | - Steven Staelens
- 3 Molecular Imaging Center Antwerp (MICA), University of Antwerp , Antwerp, Belgium
| |
Collapse
|
32
|
Juergens RA, Zukotynski KA, Singnurkar A, Snider DP, Valliant JF, Gulenchyn KY. Imaging Biomarkers in Immunotherapy. BIOMARKERS IN CANCER 2016; 8:1-13. [PMID: 26949344 PMCID: PMC4768940 DOI: 10.4137/bic.s31805] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/20/2015] [Accepted: 12/22/2015] [Indexed: 12/23/2022]
Abstract
Immune-based therapies have been in use for decades but recent work with immune checkpoint inhibitors has now changed the landscape of cancer treatment as a whole. While these advances are encouraging, clinicians still do not have a consistent biomarker they can rely on that can accurately select patients or monitor response. Molecular imaging technology provides a noninvasive mechanism to evaluate tumors and may be an ideal candidate for these purposes. This review provides an overview of the mechanism of action of varied immunotherapies and the current strategies for monitoring patients with imaging. We then describe some of the key researches in the preclinical and clinical literature on the current uses of molecular imaging of the immune system and cancer.
Collapse
Affiliation(s)
| | - Katherine A Zukotynski
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amit Singnurkar
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Denis P Snider
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - John F Valliant
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Karen Y Gulenchyn
- Department of Radiology, McMaster University, Hamilton, ON, Canada.; Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
33
|
Integration of imaging into clinical practice to assess the delivery and performance of macromolecular and nanotechnology-based oncology therapies. J Control Release 2015; 219:295-312. [PMID: 26403800 DOI: 10.1016/j.jconrel.2015.09.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/19/2015] [Accepted: 09/19/2015] [Indexed: 01/02/2023]
Abstract
Functional and molecular imaging has become increasingly used to evaluate interpatient and intrapatient tumor heterogeneity. Imaging allows for assessment of microenvironment parameters including tumor hypoxia, perfusion and proliferation, as well as tumor metabolism and the intratumoral distribution of specific molecular markers. Imaging information may be used to stratify patients for targeted therapies, and to define patient populations that may benefit from alternative therapeutic approaches. It also provides a method for non-invasive monitoring of treatment response at earlier time-points than traditional cues, such as tumor shrinkage. Further, companion diagnostic imaging techniques are becoming progressively more important for development and clinical implementation of targeted therapies. Imaging-based companion diagnostics are likely to be essential for the validation and FDA approval of targeted nanotherapies and macromolecular medicines. This review describes recent clinical advances in the use of functional and molecular imaging to evaluate the tumor microenvironment. Additionally, this article focuses on image-based assessment of distribution and anti-tumor effect of nano- and macromolecular systems.
Collapse
|
34
|
McKinley ET, Watchmaker JM, Chakravarthy AB, Meyerhardt JA, Engelman JA, Walker RC, Washington MK, Coffey RJ, Manning HC. [(18)F]-FLT PET to predict early response to neoadjuvant therapy in KRAS wild-type rectal cancer: a pilot study. Ann Nucl Med 2015; 29:535-42. [PMID: 25899481 DOI: 10.1007/s12149-015-0974-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/13/2015] [Indexed: 01/04/2023]
Abstract
OBJECT This pilot study evaluated the utility of 3'-deoxy-3'[18F]-fluorothymidine ([(18)F]-FLT) positron emission tomography (PET) to predict response to neoadjuvant therapy that included cetuximab in patients with wild-type KRAS rectal cancers. METHODS Baseline [(18)F]-FLT PET was collected prior to treatment initiation. Follow-up [(18)F]-FLT was collected after three weekly infusions of cetuximab, and following a combined regimen of cetuximab, 5-FU, and radiation. Imaging-matched biopsies were collected with each PET study. RESULTS Diminished [(18)F]-FLT PET was observed in 3/4 of patients following cetuximab treatment alone and in all patients following combination therapy. Reduced [(18)F]-FLT PET following combination therapy predicted disease-free status at surgery. Overall, [(18)F]-FLT PET agreed with Ki67 immunoreactivity from biopsy samples and surgically resected tissue, and was predictive of treatment-induced rise in p27 levels. CONCLUSION These results suggest that [(18)F]-FLT PET is a promising imaging biomarker to predict response to neoadjuvant therapy that included EGFR blockade with cetuximab in patients with rectal cancer.
Collapse
Affiliation(s)
- Eliot T McKinley
- Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical School, 1161 21st Ave. S., AA1105 MCN, Nashville, TN, 37232-2310, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Campbell BA, Callahan J, Bressel M, Simoens N, Everitt S, Hofman MS, Hicks RJ, Burbury K, MacManus M. Distribution Atlas of Proliferating Bone Marrow in Non-Small Cell Lung Cancer Patients Measured by FLT-PET/CT Imaging, With Potential Applicability in Radiation Therapy Planning. Int J Radiat Oncol Biol Phys 2015; 92:1035-1043. [PMID: 26194679 DOI: 10.1016/j.ijrobp.2015.04.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 02/03/2023]
Abstract
PURPOSE Proliferating bone marrow is exquisitely sensitive to ionizing radiation. Knowledge of its distribution could improve radiation therapy planning to minimize unnecessary marrow exposure and avoid consequential prolonged myelosuppression. [18F]-Fluoro-3-deoxy-3-L-fluorothymidine (FLT)-positron emission tomography (PET) is a novel imaging modality that provides detailed quantitative images of proliferating tissues, including bone marrow. We used FLT-PET imaging in cancer patients to produce an atlas of marrow distribution with potential clinical utility. METHODS AND MATERIALS The FLT-PET and fused CT scans of eligible patients with non-small cell lung cancer (no distant metastases, no prior cytotoxic exposure, no hematologic disorders) were reviewed. The proportions of skeletal FLT activity in 10 predefined bony regions were determined and compared according to age, sex, and recent smoking status. RESULTS Fifty-one patients were studied: 67% male; median age 68 (range, 31-87) years; 8% never smokers; 70% no smoking in the preceding 3 months. Significant differences in marrow distribution occurred between sex and age groups. No effect was detected from smoking in the preceding 3 months. Using the mean percentages of FLT uptake per body region, we created an atlas of the distribution of functional bone marrow in 4 subgroups defined by sex and age. CONCLUSIONS This atlas has potential utility for estimating the distribution of active marrow in adult cancer patients to guide radiation therapy planning. However, because of interindividual variation it should be used with caution when radiation therapy risks ablating large proportions of active marrow; in such cases, individual FLT-PET scans may be required.
Collapse
Affiliation(s)
- Belinda A Campbell
- Department of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia.
| | - Jason Callahan
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Mathias Bressel
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Nathalie Simoens
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Sarah Everitt
- Radiotherapy Services, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Michael S Hofman
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Rodney J Hicks
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Kate Burbury
- Department of Haematology, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Michael MacManus
- Department of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne, Australia
| |
Collapse
|
36
|
Abstract
Positron emission tomography (PET) is an extraordinarily sensitive clinical imaging modality for interrogating tumor metabolism. Radiolabeled PET substrates can be traced at subphysiological concentrations, allowing noninvasive imaging of metabolism and intratumoral heterogeneity in systems ranging from advanced cancer models to patients in the clinic. There are a wide range of novel and more established PET radiotracers, which can be used to investigate various aspects of the tumor, including carbohydrate, amino acid, and fatty acid metabolism. In this review, we briefly discuss the more established metabolic tracers and describe recent work on the development of new tracers. Some of the unanswered questions in tumor metabolism are considered alongside new technical developments, such as combined PET/magnetic resonance imaging scanners, which could provide new imaging solutions to some of the outstanding diagnostic challenges facing modern cancer medicine.
Collapse
Affiliation(s)
- David Y. Lewis
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Dmitry Soloviev
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|
37
|
Humbert O, Cochet A, Coudert B, Berriolo-Riedinger A, Kanoun S, Brunotte F, Fumoleau P. Role of positron emission tomography for the monitoring of response to therapy in breast cancer. Oncologist 2015; 20:94-104. [PMID: 25561512 DOI: 10.1634/theoncologist.2014-0342] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This review considers the potential utility of positron emission tomography (PET) tracers in the setting of response monitoring in breast cancer, with a special emphasis on glucose metabolic changes assessed with (18)F-fluorodeoxyglucose (FDG). In the neoadjuvant setting of breast cancer, the metabolic response can predict the final complete pathologic response after the first cycles of chemotherapy. Because tumor metabolic behavior highly depends on cancer subtype, studies are ongoing to define the optimal metabolic criteria of tumor response in each subtype. The recent multicentric randomized AVATAXHER trial has suggested, in the human epidermal growth factor 2-positive subtype, a clinical benefit of early tailoring the neoadjuvant treatment in women with poor metabolic response after the first course of treatment. In the bone-dominant metastatic setting, there is increasing clinical evidence that FDG-PET/computed tomography (CT) is the most accurate imaging modality for assessment of the tumor response to treatment when both metabolic information and morphologic information are considered. Nevertheless, there is a need to define standardized metabolic criteria of response, including the heterogeneity of response among metastases, and to evaluate the costs and health outcome of FDG-PET/CT compared with conventional imaging. New non-FDG radiotracers highlighting specific molecular hallmarks of breast cancer cells have recently emerged in preclinical and clinical studies. These biomarkers can take into account the heterogeneity of tumor biology in metastatic lesions. They may provide valuable clinical information for physicians to select and monitor the effectiveness of novel therapeutics targeting the same molecular pathways of breast tumor.
Collapse
Affiliation(s)
- Olivier Humbert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alexandre Cochet
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Bruno Coudert
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Alina Berriolo-Riedinger
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Salim Kanoun
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - François Brunotte
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| | - Pierre Fumoleau
- Departments of Nuclear Medicine and Medical Oncology, Centre G.F. Leclerc, Dijon, France; Imaging Department, Centre Hospitalo-Universitaire Le Bocage, Dijon, France; Université de Bourgogne, UMR CNRS 6306, Dijon, France
| |
Collapse
|
38
|
Yip S, Chen AB, Aerts HJWL, Berbeco R. Sensitivity study of voxel-based PET image comparison to image registration algorithms. Med Phys 2014; 41:111714. [DOI: 10.1118/1.4898125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
39
|
McKinley ET, Zhao P, Coffey RJ, Washington MK, Manning HC. 3'-Deoxy-3'-[18F]-Fluorothymidine PET imaging reflects PI3K-mTOR-mediated pro-survival response to targeted therapy in colorectal cancer. PLoS One 2014; 9:e108193. [PMID: 25247710 PMCID: PMC4172755 DOI: 10.1371/journal.pone.0108193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 08/24/2014] [Indexed: 01/02/2023] Open
Abstract
Biomarkers that predict response to targeted therapy in oncology are an essential component of personalized medicine. In preclinical treatment response studies that featured models of wild-type KRAS or mutant BRAF colorectal cancer treated with either cetuximab or vemurafenib, respectively, we illustrate that [18F]-FLT PET, a non-invasive molecular imaging readout of thymidine salvage, closely reflects pro-survival responses to targeted therapy that are mediated by PI3K-mTOR activity. Activation of pro-survival mechanisms forms the basis of numerous modes of resistance. Therefore, we conclude that [18F]-FLT PET may serve a novel and potentially critical role to predict tumors that exhibit molecular features that tend to reflect recalcitrance to MAPK-targeted therapy. Though these studies focused on colorectal cancer, we envision that the results may be applicable to other solid tumors as well.
Collapse
Affiliation(s)
- Eliot T. McKinley
- The Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Biomedical Engineering, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN, United States of America
| | - Ping Zhao
- The Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical School, Nashville, TN, United States of America
| | - Robert J. Coffey
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Vanderbilt Ingram Cancer Center, Vanderbilt University Medical School, Nashville, TN, United States of America
| | - M. Kay Washington
- Department of Medicine, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Vanderbilt Ingram Cancer Center, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Pathology, Vanderbilt University Medical School, Nashville, TN, United States of America
| | - H. Charles Manning
- The Vanderbilt University Institute of Imaging Science (VUIIS), Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Biomedical Engineering, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Vanderbilt Ingram Cancer Center, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Neurosurgery, Vanderbilt University Medical School, Nashville, TN, United States of America
- Department of Chemical and Physical Biology, Vanderbilt University Medical School, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
40
|
Methodological considerations in quantification of 3'-deoxy-3'-[18F]fluorothymidine uptake measured with positron emission tomography in patients with non-small cell lung cancer. Mol Imaging Biol 2014; 16:136-45. [PMID: 23813332 DOI: 10.1007/s11307-013-0658-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE To investigate the effect of image-derived input functions (IDIF), input function corrections and volume of interest (VOI) size in quantification of [(18)F]FLT uptake in non-small cell lung cancer (NSCLC) patients. PROCEDURES Twenty-three NSCLC patients were scanned on a HR+ scanner. IDIFs were defined over the aorta and left ventricle. Activity concentration and metabolite fraction were measured in venous blood samples. Venous blood samples at 30, 40 and 60 min after injection were used to calibrate the IDIF time-activity curves. Adaptive thresholds were used for VOI definition. Full kinetic analysis and simplified measures were performed. RESULTS Non-linear regression analysis showed better fits for the irreversible model compared to the reversible model in the majority. Calibrated and metabolite corrected plus plasma-to-blood ratio corrected input function resulted in high correlations between SUV and Patlak K i (Pearson correlation coefficients 0.86-0.96, p value < 0.001). No significant differences in correlation between SUV and Patlak K i were observed with variation of IDIF structure or VOI size. CONCLUSIONS Plasma-to-blood ratio correction, metabolite correction and calibration improved the correlation between SUV and Patlak K i significantly, indicating the need for these corrections when K i is used to validate semi-quantitative measures, such as SUV.
Collapse
|
41
|
Yang J, Yue JB, Liu J, Yu JM. Repopulation of tumor cells during fractionated radiotherapy and detection methods (Review). Oncol Lett 2014; 7:1755-1760. [PMID: 24932228 PMCID: PMC4049693 DOI: 10.3892/ol.2014.1990] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 02/26/2014] [Indexed: 12/18/2022] Open
Abstract
Repopulation of tumor cells during radiotherapy is believed to be a significant cause for treatment failure. The phenomenon of tumor repopulation during fractionated radiotherapy was found from clinical observations that identified that the local control rate decreased with a prolonged treatment time. A series of animal experiments with varied overall treatment time and fractionated doses were performed to demonstrate tumor cell repopulation during radiotherapy in various mouse xenograft models. However, conventional detection methods are challenging, as it is difficult to separate viable cells from those destined for apoptosis during fractionated radiotherapy. In essence, the mechanism of tumor repopulation involves the continuing proliferation of clonogenic tumor cells. In vivo imaging, tracking and targeting of the repopulation of these cells has been of clinical interest so as to administer a higher dose to the tumor repopulation regions. Currently, functional imaging methods, including 3'-deoxy-3'-18F-fluorothymidine positron emission tomography (18F-FLT PET), are showing promise in assessing the proliferation activity of tumors in vivo. This review mainly focuses on the phenomenon of tumor repopulation during radiotherapy and its conventional and novel detection methods, particularly on the feasibility of 18F-FLT PET for the detection of tumor-cell repopulation.
Collapse
Affiliation(s)
- Jia Yang
- Shandong Cancer Hospital and Institute, Jinan University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jin-Bo Yue
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Jing Liu
- Graduate Education Centre of Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Jin-Ming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
42
|
Bardram Johnbeck C, Munk Jensen M, Haagen Nielsen C, Fisker Hag AM, Knigge U, Kjaer A. 18F-FDG and 18F-FLT-PET imaging for monitoring everolimus effect on tumor-growth in neuroendocrine tumors: studies in human tumor xenografts in mice. PLoS One 2014; 9:e91387. [PMID: 24626055 PMCID: PMC3953383 DOI: 10.1371/journal.pone.0091387] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 02/08/2014] [Indexed: 12/31/2022] Open
Abstract
Introduction The mTOR inhibitor everolimus has shown promising results in some but not all neuroendocrine tumors. Therefore, early assessment of treatment response would be beneficial. In this study, we investigated the in vivo and in vitro treatment effect of everolimus in neuroendocrine tumors and evaluated the performance of 18F-FDG and the proliferation tracer 18F-FLT for treatment response assessment by PET imaging. Methods The effect of everolimus on the human carcinoid cell line H727 was examined in vitro with the MTT assay and in vivo on H727 xenograft tumors. The mice were scanned at baseline with 18F-FDG or 18F-FLT and then treated with either placebo or everolimus (5 mg/kg daily) for 10 days. PET/CT scans were repeated at day 1,3 and 10. Results Everolimus showed significant inhibition of H727 cell proliferation in vitro at concentrations above 1 nM. In vivo tumor volumes measured relative to baseline were significantly lower in the everolimus group compared to the control group at day 3 (126±6% vs. 152±6%; p = 0.016), day 7 (164±7% vs. 226±13%; p<0.001) and at day 10 (194±10% vs. 281±18%; p<0.001). Uptake of 18F-FDG and 18F-FLT showed little differences between control and treatment groups, but individual mean uptake of 18F-FDG at day 3 correlated with tumor growth day 10 (r2 = 0.45; P = 0.034), 18F-FLT mean uptake at day 1 correlated with tumor growth day 7 (r2 = 0.63; P = 0.019) and at day 3 18F-FLT correlated with tumor growth day 7 (r2 = 0.87; P<0.001) and day 10 (r2 = 0.58; P = 0.027). Conclusion Everolimus was effective in vitro and in vivo in human xenografts lung carcinoid NETs and especially early 18F-FLT uptake predicted subsequent tumor growth. We suggest that 18F-FLT PET can be used for tailoring therapy for neuroendocrine tumor patients through early identification of responders and non-responders.
Collapse
Affiliation(s)
- Camilla Bardram Johnbeck
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Faculty of Health Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Mette Munk Jensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Faculty of Health Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Carsten Haagen Nielsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Faculty of Health Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Anne Mette Fisker Hag
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Faculty of Health Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Knigge
- Department of Surgical Gastroenterology C and Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Faculty of Health Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Lin G, Chung YL. Current opportunities and challenges of magnetic resonance spectroscopy, positron emission tomography, and mass spectrometry imaging for mapping cancer metabolism in vivo. BIOMED RESEARCH INTERNATIONAL 2014; 2014:625095. [PMID: 24724090 PMCID: PMC3958648 DOI: 10.1155/2014/625095] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/06/2014] [Accepted: 01/19/2014] [Indexed: 12/18/2022]
Abstract
Cancer is known to have unique metabolic features such as Warburg effect. Current cancer therapy has moved forward from cytotoxic treatment to personalized, targeted therapies, with some that could lead to specific metabolic changes, potentially monitored by imaging methods. In this paper we addressed the important aspects to study cancer metabolism by using image techniques, focusing on opportunities and challenges of magnetic resonance spectroscopy (MRS), dynamic nuclear polarization (DNP)-MRS, positron emission tomography (PET), and mass spectrometry imaging (MSI) for mapping cancer metabolism. Finally, we highlighted the future possibilities of an integrated in vivo PET/MR imaging systems, together with an in situ MSI tissue analytical platform, may become the ultimate technologies for unraveling and understanding the molecular complexities in some aspects of cancer metabolism. Such comprehensive imaging investigations might provide information on pharmacometabolomics, biomarker discovery, and disease diagnosis, prognosis, and treatment response monitoring for clinical medicine.
Collapse
Affiliation(s)
- Gigin Lin
- Department of Radiology, Chang Gung Memorial Hospital at Linkou, Chang Gung University, 5 Fuhsing Street, Guishan, Taoyuan 333, Taiwan
- Molecular Imaging Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University, 5 Fuhsing Street, Guishan, Taoyuan 333, Taiwan
- Metabolomics Core Laboratory, Chang Gung Memorial Hospital at Linkou, Chang Gung University, 5 Fuhsing Street, Guishan, Taoyuan 333, Taiwan
| | - Yuen-Li Chung
- The Institute of Cancer Research and Royal Marsden Hospital, CRUK Cancer Imaging Centre, Downs Road, Sutton, Surrey SM2 5PT, UK
| |
Collapse
|
44
|
Thymidine phosphorylase influences [18F]fluorothymidine uptake in cancer cells and patients with non-small cell lung cancer. Eur J Nucl Med Mol Imaging 2014; 41:1327-35. [DOI: 10.1007/s00259-014-2712-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 01/20/2014] [Indexed: 01/09/2023]
|
45
|
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]
|
46
|
Cunha L, Szigeti K, Mathé D, Metello LF. The role of molecular imaging in modern drug development. Drug Discov Today 2014; 19:936-48. [PMID: 24434047 DOI: 10.1016/j.drudis.2014.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 11/30/2013] [Accepted: 01/07/2014] [Indexed: 12/28/2022]
Abstract
Drug development represents a highly complex, inefficient and costly process. Over the past decade, the widespread use of nuclear imaging, owing to its functional and molecular nature, has proven to be a determinant in improving the efficiency in selecting the candidate drugs that should either be abandoned or moved forward into clinical trials. This helps not only with the development of safer and effective drugs but also with the shortening of time-to-market. The modern concept and future trends concerning molecular imaging will assumedly be hybrid or multimodality imaging, including combinations between high sensitivity and functional (molecular) modalities with high spatial resolution and morphological techniques.
Collapse
Affiliation(s)
- Lídia Cunha
- Nuclear Medicine Department, High Institute for Allied Health Technologies, Polytechnic Institute of Porto (ESTSP.IPP), Vila Nova de Gaia 4400-330, Portugal
| | - Krisztián Szigeti
- Nanobiotechnology &In Vivo Imaging Center, Semmelweis University, Budapest H-1094, Hungary
| | - Domokos Mathé
- CROmed Ltd, H-1047 Budapest Baross u. 91-95, Budapest, Hungary
| | - Luís F Metello
- Nuclear Medicine Department, High Institute for Allied Health Technologies, Polytechnic Institute of Porto (ESTSP.IPP), Vila Nova de Gaia 4400-330, Portugal; IsoPor, SA, Porto, Portugal.
| |
Collapse
|
47
|
Wondergem MJ, Herrmann K, Syrbu S, Zijlstra JM, Hoetjes N, Hoekstra OS, Cillessen SA, Moesbergen LM, Buck AK, Vose JM, Juweid ME. 18 F-fluorothymidine uptake in follicular lymphoma and error-prone DNA repair. EJNMMI Res 2014; 4:3. [PMID: 24397937 PMCID: PMC3895783 DOI: 10.1186/2191-219x-4-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/18/2013] [Indexed: 11/23/2022] Open
Abstract
Background We observed a disproportional 18 F-fluorothymidine (F-FLT) uptake in follicular lymphoma (FL) relative to its low cell proliferation. We tested the hypothesis that the ‘excess’ uptake of 18 F-FLT in FL is related to error-prone DNA repair and investigated whether this also contributes to 18 F-FLT uptake in diffuse large B cell lymphoma (DLBCL). Methods We performed immunohistochemical stainings to assess the pure DNA replication marker MIB-1 as well as markers of both DNA replication and repair like PCNA, TK-1 and RPA1 on lymph node biopsies of 27 FLs and 35 DLBCLs. In 7 FL and 15 DLBCL patients, 18 F-FLT-PET had been performed. Results 18 F-FLT uptake was lower in FL than in DLBCL (median SUVmax 5.7 vs. 8.9, p = 0,004), but the ratio of 18 F-FLT-SUVmax to percentage of MIB-1 positive cells was significantly higher in FL compared with DLBCL (p = 0.001). The median percentage of MIB-1 positive cells was 10% (range, 10% to 20%) in FL and 70% (40% to 80%) in DLBCL. In contrast, the median percentages of PCNA, TK-1 and RPA1 positive cells were 90% (range, 80 to 100), 90% (80 to 100) and 100% (80 to 100) in FL versus 90% (60 to 100), 90% (60 to 100) and 100% (80 to 100) in DLBCL, respectively. Conclusions This is the first demonstration of a striking discordance between 18 F-FLT uptake in FL and tumour cell proliferation. High expression of DNA replication and repair markers compared with the pure proliferation marker MIB-1 in FL suggests that this discordance might be due to error-prone DNA repair. While DNA repair-related 18 F-FLT uptake considerably contributes to 18 F-FLT uptake in FL, its contribution to 18 F-FLT uptake in highly proliferative DLBCL is small. This apparently high contribution of DNA repair to the 18 F-FLT signal in FL may hamper studies where 18 F-FLT is used to assess response to cytostatic therapy or to distinguish between FL and transformed lymphoma.
Collapse
Affiliation(s)
- Marielle J Wondergem
- Department of Haematology, VU University Medical Center (VUMC), De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Keen HG, Ricketts SA, Maynard J, Logie A, Odedra R, Shannon AM, Wedge SR, Guichard SM. Examining Changes in [18 F]FDG and [18 F]FLT Uptake in U87-MG Glioma Xenografts as Early Response Biomarkers to Treatment with the Dual mTOR1/2 Inhibitor AZD8055. Mol Imaging Biol 2013; 16:421-30. [DOI: 10.1007/s11307-013-0705-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Karroum O, Mignion L, Kengen J, Karmani L, Levêque P, Danhier P, Magat J, Bol A, Labar D, Grégoire V, Bouzin C, Feron O, Gallez B, Jordan BF. Multimodal imaging of tumor response to sorafenib combined with radiation therapy: comparison between diffusion-weighted MRI, choline spectroscopy and 18F-FLT PET imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:274-80. [PMID: 23606431 DOI: 10.1002/cmmi.1525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 11/08/2012] [Accepted: 11/29/2012] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to determine the value of different imaging modalities, that is, magnetic resonance imaging/spectroscopy (MRI/MRS) and positron emission tomography (PET), to assess early tumor response to sorafenib with or without radiotherapy. Diffusion-weighted (DW)-MRI, choline (1)H MRS at 11.7 T, and (18)F-FLT PET imaging were used to image fibrosarcoma (FSaII) tumor-bearing mice over time. The imaging markers were compared with apoptosis cell death and cell proliferation measurements assessed by histology. Anti-proliferative effects of sorafenib were evidenced by (1)H MRS and (18)F-FLT PET after 2 days of treatment with sorafenib, with no additional effect of the combination with radiation therapy, results that are in agreement with Ki67 staining. Apparent diffusion coefficient calculated using DW-MRI was not modified after 2 days of treatment with sorafenib, but showed significant increase 24 h after 2 days of sorafenib treatment combined with consecutive irradiation. The three imaging markers were able to show early tumor response as soon as 24 h after treatment initiation, with choline MRS and (18)F-FLT being sensitive to sorafenib in monotherapy as well as in combined therapy with irradiation, whereas DW-MRI was only sensitive to the combination of sorafenib with radiotherapy.
Collapse
Affiliation(s)
- Oussama Karroum
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain, Belgium, Avenue Mounier 73, B-1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 PMCID: PMC3799232 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|