1
|
Lasnon C, Morel A, Aide N, Silva AD, Emile G. Baseline and early 18F-FDG PET/CT evaluations as predictors of progression-free survival in metastatic breast cancer patients treated with targeted anti-CDK therapy. Cancer Imaging 2024; 24:90. [PMID: 38982546 PMCID: PMC11232230 DOI: 10.1186/s40644-024-00727-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 06/20/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Exploring the value of baseline and early 18F-FDG PET/CT evaluations in prediction PFS in ER+/HER2- metastatic breast cancer patients treated with a cyclin-dependent kinase inhibitor in combination with an endocrine therapy. METHODS Sixty-six consecutive breast cancer patients who underwent a pre-therapeutic 18F-FDG PET/CT and a second PET/CT within the first 6 months of treatment were retrospectively included. Metabolic tumour volume (MTV) and total lesion glycolysis (TLG) and Dmax, which represents tumour dissemination and is defined as the distance between the two most distant lesions, were computed. The variation in these parameters between baseline and early evaluation PET as well as therapeutic evaluation using PERCIST were assessed as prognosticators of PFS at 18 months. RESULTS The median follow-up was equal to 22.5 months. Thirty progressions occurred (45.4%). The average time to event was 17.8 ± 10.4 months. At baseline, Dmax was the only predictive metabolic parameter. Patients with a baseline Dmax ≤ 18.10 cm had a significantly better 18 m-PFS survival than the others: 69.2% (7.7%) versus 36.7% (8.8%), p = 0.017. There was no association between PERCIST evaluation and 18 m-PFS status (p = 0.149) and there was no difference in 18 m-PFS status between patients classified as complete, partial metabolic responders or having stable metabolic disease. CONCLUSION Disease spread at baseline PET, as assessed by Dmax, is predictive of an event occurring within 18 months. In the absence of early metabolic progression, which occurs in 15% of patients, treatment should be continued regardless of the quality of the initial response to treatment.
Collapse
Affiliation(s)
- Charline Lasnon
- Nuclear Medicine Department, François Baclesse Comprehensive Cancer Center, UNICANCER, 3 Avenue du General Harris, BP 45026, Caen Cedex 5, 14076, France.
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, Caen, France.
| | - Adeline Morel
- Medical Oncology Department, François Baclesse Comprehensive Cancer Center, UNICANCER, Caen, France
| | - Nicolas Aide
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, Caen, France
| | - Angélique Da Silva
- Medical Oncology Department, François Baclesse Comprehensive Cancer Center, UNICANCER, Caen, France
| | - George Emile
- Medical Oncology Department, François Baclesse Comprehensive Cancer Center, UNICANCER, Caen, France
| |
Collapse
|
2
|
Jadhav PV, Prasath NJ, Gajbhiye SG, Rane UA, Agnihotri TG, Gomte SS, Jain A. Empowering the Battle: Bioenhancers as Allies Against Cancer Drug Resistance. Curr Pharm Biotechnol 2024; 25:1552-1563. [PMID: 37957922 DOI: 10.2174/0113892010192038231107051715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Drug resistance has been a great hindrance in the path of counteracting diseases like cancer and is driven by drugs misuse and overuse. In terms of cancer, resistance has been developed due to cellular changes, altered growth activation pathways, increased expression of efflux proteins, and changes in the local physiology of cancer (blood supply, tissue hydrodynamics, increased mutation rate/epigenetics, tumor cell heterogeneity). One of the approaches to address these challenges is the use of bioenhancers, which can overcome drug resistance, thereby improving bioavailability (BA). CONCLUSION Bioenhancers when combined with drugs can elicit pharmacological activity. They are generally combined with therapeutic agents at low doses, which increase the BA or therapeutic activity of active pharmaceutical ingredient (API). This review sheds light on the synthesis and classification of bio-enhancers. It also discusses different applications of bio-enhancers like piperine, ginger, quercetin, curcumin, etc. in the treatment of cancer. The review also presents some of the recent advancements in terms of nanocarriers for delivering API combined with bioenhancers.
Collapse
Affiliation(s)
- Pratiksha Vasant Jadhav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Naga Jothi Prasath
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Saurabh Ghannil Gajbhiye
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Utkarsha Arun Rane
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research- Ahmedabad, Palaj, Opposite to Air Force Station, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
3
|
Schwenck J, Sonanini D, Cotton JM, Rammensee HG, la Fougère C, Zender L, Pichler BJ. Advances in PET imaging of cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00576-4. [PMID: 37258875 DOI: 10.1038/s41568-023-00576-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Molecular imaging has experienced enormous advancements in the areas of imaging technology, imaging probe and contrast development, and data quality, as well as machine learning-based data analysis. Positron emission tomography (PET) and its combination with computed tomography (CT) or magnetic resonance imaging (MRI) as a multimodality PET-CT or PET-MRI system offer a wealth of molecular, functional and morphological data with a single patient scan. Despite the recent technical advances and the availability of dozens of disease-specific contrast and imaging probes, only a few parameters, such as tumour size or the mean tracer uptake, are used for the evaluation of images in clinical practice. Multiparametric in vivo imaging data not only are highly quantitative but also can provide invaluable information about pathophysiology, receptor expression, metabolism, or morphological and functional features of tumours, such as pH, oxygenation or tissue density, as well as pharmacodynamic properties of drugs, to measure drug response with a contrast agent. It can further quantitatively map and spatially resolve the intertumoural and intratumoural heterogeneity, providing insights into tumour vulnerabilities for target-specific therapeutic interventions. Failure to exploit and integrate the full potential of such powerful imaging data may lead to a lost opportunity in which patients do not receive the best possible care. With the desire to implement personalized medicine in the cancer clinic, the full comprehensive diagnostic power of multiplexed imaging should be utilized.
Collapse
Affiliation(s)
- Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonathan M Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Department of Immunology, IFIZ Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany.
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
4
|
18F-fluorodeoxyglucose positron emission tomography correlates with tumor immunometabolic phenotypes in resected lung cancer. Cancer Immunol Immunother 2020; 69:1519-1534. [PMID: 32300858 DOI: 10.1007/s00262-020-02560-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Enhanced tumor glycolytic activity is a mechanism by which tumors induce an immunosuppressive environment to resist adoptive T cell therapy; therefore, methods of assessing intratumoral glycolytic activity are of considerable clinical interest. In this study, we characterized the relationships among tumor 18F-fluorodeoxyglucose (FDG) retention, tumor metabolic and immune phenotypes, and survival in patients with resected non-small cell lung cancer (NSCLC). We retrospectively analyzed tumor preoperative positron emission tomography (PET) 18F-FDG uptake in 59 resected NSCLCs and investigated correlations between PET parameters (SUVMax, SUVTotal, SUVMean, TLG), tumor expression of glycolysis- and immune-related genes, and tumor-associated immune cell densities that were quantified by immunohistochemistry. Tumor glycolysis-associated immune gene signatures were analyzed for associations with survival outcomes. We found that each 18F-FDG PET parameter was positively correlated with tumor expression of glycolysis-related genes. Elevated 18F-FDG SUVMax was more discriminatory of glycolysis-associated changes in tumor immune phenotypes than other 18F-FDG PET parameters. Increased SUVMax was associated with multiple immune factors characteristic of an immunosuppressive and poorly immune infiltrated tumor microenvironment, including elevated PD-L1 expression, reduced CD57+ cell density, and increased T cell exhaustion gene signature. Elevated SUVMax identified immune-related transcriptomic signatures that were associated with enhanced tumor glycolytic gene expression and poor clinical outcomes. Our results suggest that 18F-FDG SUVMax has potential value as a noninvasive, clinical indicator of tumor immunometabolic phenotypes in patients with resectable NSCLC and warrants investigation as a potential predictor of therapeutic response to immune-based treatment strategies.
Collapse
|
5
|
Serrano C, Leal A, Kuang Y, Morgan JA, Barysauskas CM, Phallen J, Triplett O, Mariño-Enríquez A, Wagner AJ, Demetri GD, Velculescu VE, Paweletz CP, Fletcher JA, George S. Phase I Study of Rapid Alternation of Sunitinib and Regorafenib for the Treatment of Tyrosine Kinase Inhibitor Refractory Gastrointestinal Stromal Tumors. Clin Cancer Res 2019; 25:7287-7293. [PMID: 31471313 DOI: 10.1158/1078-0432.ccr-19-2150] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/05/2019] [Accepted: 08/27/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE Polyclonal emergence of KIT secondary mutations is a main mechanism of imatinib progression in gastrointestinal stromal tumor (GIST). Approved KIT inhibitors sunitinib and regorafenib have complementary activity against KIT resistance mutations. Preclinical evidence suggests that rapid alternation of sunitinib and regorafenib broadens the spectrum of imatinib-resistant subclones targeted. PATIENTS AND METHODS Phase Ib study investigating continuous treatment with cycles of sunitinib (3 days) followed by regorafenib (4 days) in patients with tyrosine kinase inhibitor (TKI)-refractory GIST. A 3+3 dosing schema was utilized to determine the recommended phase II dose (RP2D). Plasma samples were analyzed for pharmacokinetics and circulating tumor DNA (ctDNA) studies using targeted error correction sequencing (TEC-seq) and droplet digital PCR (ddPCR). RESULTS Of the 14 patients enrolled, 2 experienced dose-limiting toxicities at dose level 2 (asymptomatic grade 3 hypophosphatemia). Sunitinib 37.5 mg/day and regorafenib 120 mg/day was the RP2D. Treatment was well-tolerated and no unexpected toxicities resulted from the combination. Stable disease was the best response in 4 patients, and median progression-free survival was 1.9 months. Combined assessment of ctDNA with TEC-seq and ddPCR detected plasma mutations in 11 of 12 patients (92%). ctDNA studies showed that KIT secondary mutations remain the main mechanism of resistance in TKI-refractory GIST, revealing effective suppression of KIT-mutant subpopulations in patients benefiting from the combination. CONCLUSIONS Sunitinib and regorafenib combination is feasible and tolerable. Rapid alternation of TKIs with complementary activity might be effective when combining drugs with favorable pharmacokinetics, potentially allowing active doses while minimizing adverse events. Serial monitoring with ctDNA may guide treatment in patients with GIST.
Collapse
Affiliation(s)
- César Serrano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts.,Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology; Department of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Alessandro Leal
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yanan Kuang
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jeffrey A Morgan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Olivia Triplett
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adrián Mariño-Enríquez
- Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Andrew J Wagner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - George D Demetri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Ludwig Center for Cancer Research at Dana-Farber Cancer Institute and Harvard Medical School, Boston Massachusetts
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cloud P Paweletz
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | - Suzanne George
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
6
|
Taralli S, Lorusso M, Scolozzi V, Masiello V, Marazzi F, Calcagni ML. Response evaluation with 18F-FDG PET/CT in metastatic breast cancer patients treated with Palbociclib: first experience in clinical practice. Ann Nucl Med 2018; 33:193-200. [PMID: 30569442 DOI: 10.1007/s12149-018-01323-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/02/2018] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Palbociclib is a cyclin-dependent kinase 4/6 inhibitor recently approved for treatment in advanced or metastatic breast cancer (BC) patients. The use of 18F-FDG PET/CT for chemo/endocrine therapy response assessment in BC patients is well reported in the literature, but no studies have evaluated its role for assessing Palbociclib efficacy in clinical practice. Our study aimed to evaluate the potential role of 18F-FDG PET/CT in this setting. METHODS In 12 metastatic BC patients (mean age = 62 ± 10 years) treated with Palbociclib plus endocrine therapy and who underwent a baseline and post-therapy 18F-FDG PET/CT, we retrospectively compared the Metabolic Response Evaluation (MRE, based on PET/CT) to the Standard Response Evaluation (SRE, based on clinico-laboratory and morphological data); we also assessed the influence of additional PET/CT information on the patients' management. RESULTS Compared to SRE, MRE increased the proportion of patients classified with progressive disease from 25 to 50% and differed from SRE in 8/12 patients: 3/8 shifted from stable disease or undetermined response to metabolic progression (more unfavorable category), 4/8 from stable disease to partial or complete metabolic response, and 1/8 from partial response to complete metabolic response (more favorable category). Additional PET/CT information led to a change in patients' management in 3/12 (25%) patients. CONCLUSION In BC patients treated with Palbociclib, additional 18F-FDG PET/CT information seems clinically useful, with respect to personalized management, to early intercept patients who should discontinue Palbociclib because of progressive disease and to select patients requiring a strict monitoring of additional metabolic findings. Further studies are needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Silvia Taralli
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito, 1, 00168, Rome, Italy.
| | - Margherita Lorusso
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito, 1, 00168, Rome, Italy
| | - Valentina Scolozzi
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito, 1, 00168, Rome, Italy.,Nuclear Medicine Institute, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valeria Masiello
- Radiation Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fabio Marazzi
- Radiation Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Lucia Calcagni
- Nuclear Medicine Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Francesco Vito, 1, 00168, Rome, Italy.,Nuclear Medicine Institute, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Höink A, Persigehl T, Kwiecien R, Balthasar M, Mesters R, Berdel W, Heindel W, Bremer C, Schwöppe C. Gadofosveset-enhanced MRI as simple surrogate parameter for real-time evaluation of the initial tumour vessel infarction by retargeted tissue factor tTF-NGR. Oncol Lett 2018; 17:270-280. [PMID: 30655764 PMCID: PMC6313167 DOI: 10.3892/ol.2018.9638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/22/2018] [Indexed: 01/16/2023] Open
Abstract
Truncated tissue factor (tTF)-NGR consists of the extracellular domain of the human TF and the binding motif NGR. tTF-NGR activates blood coagulation within the tumour vasculature following binding to CD13, and is overexpressed in the endothelial cells of tumour vessels, resulting in tumour vessel infarction and subsequent retardation/regression of tumour growth. The aim of the present study was to investigate gadofosveset-based real-time dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in evaluating the initial therapeutic effects of the anti-vascular tTF-NGR approach. DCE-MRI (3.0 T) was performed in human U87-glioblastoma tumour-bearing nude mice. During a dynamic T1w GE-sequence, a gadolinium-based blood pool contrast agent (gadofosveset) was injected via a tail vein catheter. Following the maximum contrast intensity inside the tumour being obtained, tTF-NGR was injected (controls received NaCl) and the contrast behaviour of the tumour was monitored by ROI analysis. The slope difference of signal intensities between controls and the tTF-NGR group was investigated, as well as the differences between the average area under the curve (AUC) of the two groups. The association between intensity, group (control vs. tTF-NGR group) and time was analysed by fitting a linear mixed model. Following the injection of tTF-NGR, the signal intensity inside the tumours exhibited a statistically significantly stronger average slope decrease compared with the signal intensity of the tumours in the NaCl group. Furthermore, the initial average AUC values of mice treated with tTF-NGR were 5.7% lower than the average AUC of the control animals (P<0.05). Gadofosveset-enhanced MRI enables the visualization of the initial tumour response to anti-vascular treatment in real-time. Considering the clinical application of tTF-NGR, this method may provide a simple alternative parameter for monitoring the tumour response to vascular disrupting agents and certain vascular targeting agents in humans.
Collapse
Affiliation(s)
- Anna Höink
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Thorsten Persigehl
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Robert Kwiecien
- Institute of Biostatistics and Clinical Research, University of Münster, Germany
| | - Martin Balthasar
- Department of Diagnostic and Interventional Radiology, University Hospital Cologne, D-50937 Cologne, Germany
| | - Rolf Mesters
- Department of Medicine A - Haematology and Oncology, University Hospital Münster, D-48149 Münster, Germany
| | - Wolfgang Berdel
- Department of Medicine A - Haematology and Oncology, University Hospital Münster, D-48149 Münster, Germany
| | - Walter Heindel
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany
| | - Christoph Bremer
- Department of Clinical Radiology, University Hospital Münster, D-48149 Münster, Germany.,Department of Radiology, St. Franziskus-Hospital Münster, D-48145 Münster, Germany
| | - Christian Schwöppe
- Department of Radiology, St. Franziskus-Hospital Münster, D-48145 Münster, Germany
| |
Collapse
|
8
|
Aung W, Tsuji AB, Sudo H, Sugyo A, Ukai Y, Kouda K, Kurosawa Y, Furukawa T, Saga T. Radioimmunotherapy of pancreatic cancer xenografts in nude mice using 90Y-labeled anti-α6β4 integrin antibody. Oncotarget 2018; 7:38835-38844. [PMID: 27246980 PMCID: PMC5122433 DOI: 10.18632/oncotarget.9631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/04/2016] [Indexed: 12/31/2022] Open
Abstract
The contribution of integrin α6β4 (α6β4) overexpression to the pancreatic cancer invasion and metastasis has been previously shown. We have reported immunotargeting of α6β4 for radionuclide-based and near-infrared fluorescence imaging in a pancreatic cancer model. In this study, we prepared yttrium-90 labeled anti-α6β4 antibody (90Y-ITGA6B4) and evaluated its radioimmunotherapeutic efficacy against pancreatic cancer xenografts in nude mice. Mice bearing xenograft tumors were randomly divided into 5 groups: (1) single administration of 90Y-ITGA6B4 (3.7MBq), (2) double administrations of 90Y-ITGA6B4 with once-weekly schedule (3.7MBq × 2), (3) single administration of unlabeled ITGA6B4, (4) double administrations of unlabeled ITGA6B4 with once-weekly schedule and (5) the untreated control. Biweekly tumor volume measurements and immunohistochemical analyses of tumors at 2 days post-administration were performed to monitor the response to treatments. To assess the toxicity, body weight was measured biweekly. Additionally, at 27 days post-administration, blood samples were collected through cardiac puncture, and hematological parameters, hepatic and renal functions were analyzed. Both 90Y-ITGA6B4 treatment groups showed reduction in tumor volumes (P < 0.04), decreased cell proliferation marker Ki-67-positive cells and increased DNA damage marker p-H2AX-positive cells, compared with the other groups. Mice treated with double administrations of 90Y-ITGA6B4, exhibited myelosuppression. There were no significant differences in hepatic and renal functions between the 2 treatment groups and the other groups. Our results suggest that 90Y-ITGA6B4 is a promising radioimmunotherapeutic agent against α6β4 overexpressing tumors. In the future studies, dose adjustment for fractionated RIT should be considered carefully in order to get the optimal effect while avoiding myelotoxicity.
Collapse
Affiliation(s)
- Winn Aung
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Atsushi B Tsuji
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hitomi Sudo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Aya Sugyo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | | | | | - Yoshikazu Kurosawa
- Innovation Center for Advanced Medicine, Fujita Health University, Toyoake, Japan
| | - Takako Furukawa
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
9
|
Lavezzi SM, Borella E, Carrara L, De Nicolao G, Magni P, Poggesi I. Mathematical modeling of efficacy and safety for anticancer drugs clinical development. Expert Opin Drug Discov 2017; 13:5-21. [DOI: 10.1080/17460441.2018.1388369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Silvia Maria Lavezzi
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Elisa Borella
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Letizia Carrara
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Giuseppe De Nicolao
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Paolo Magni
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Italo Poggesi
- Global Clinical Pharmacology, Janssen Research and Development, Cologno Monzese, Italy
| |
Collapse
|
10
|
Molecularly targeted therapies in cancer: a guide for the nuclear medicine physician. Eur J Nucl Med Mol Imaging 2017; 44:41-54. [PMID: 28396911 PMCID: PMC5541087 DOI: 10.1007/s00259-017-3695-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 01/01/2023]
Abstract
Molecular imaging continues to influence every aspect of cancer care including detection, diagnosis, staging and therapy response assessment. Recent advances in the understanding of cancer biology have prompted the introduction of new targeted therapy approaches. Precision medicine in oncology has led to rapid advances and novel approaches optimizing the use of imaging modalities in cancer care, research and development. This article focuses on the concept of targeted therapy in cancer and the challenges that exist for molecular imaging in cancer care.
Collapse
|
11
|
Obrzut S, McCammack K, Badran KW, Balistreri A, Ou E, Nguyen BJ, Hoh CK, Rose SC. Prognostic value of post-Yttrium 90 radioembolization therapy 18F-fluorodeoxyglucose positron emission tomography in patients with liver tumors. Clin Imaging 2017; 42:43-49. [DOI: 10.1016/j.clinimag.2016.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/31/2016] [Accepted: 11/15/2016] [Indexed: 12/23/2022]
|
12
|
Mudd SR, Comley RA, Bergstrom M, Holen KD, Luo Y, Carme S, Fox GB, Martarello L, Beaver JD. Molecular imaging in oncology drug development. Drug Discov Today 2017; 22:140-147. [DOI: 10.1016/j.drudis.2016.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 08/16/2016] [Accepted: 09/21/2016] [Indexed: 01/08/2023]
|
13
|
dos Anjos RF, dos Anjos DA, Vieira DL, Leite AF, Figueiredo PTDS, de Melo NS. Effectiveness of FDG-PET/CT for evaluating early response to induction chemotherapy in head and neck squamous cell carcinoma: A systematic review. Medicine (Baltimore) 2016; 95:e4450. [PMID: 27512861 PMCID: PMC4985316 DOI: 10.1097/md.0000000000004450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND F-Fluoro-Deoxy-Glucose Positron Emission Tomography with Computed Tomography (F-FDG PET/CT) may be a powerful tool to predict treatment outcome. We aimed to review the effectiveness of F-FDG PET/CT in the assessment of early response to induction chemotherapy (IC) in patients with advanced Head and Neck Squamous Cell Cancer (HNSCC) without previous treatment. METHODS PubMed, Cochrane Library, Science Direct and Web of Science were searched to May 2016. Reference lists of the included articles and additional studies identified by one nuclear medicine expert were screened for potential relevant studies that investigated the effectiveness of F-FDG PET/CT performed before and after IC. Three authors independently screened all retrieved articles, selected studies that met inclusion criteria and extracted data. The methodology of the selected studies was evaluated by using the risk of bias checklist of the Agency for Healthcare Research and Quality (AHRQ). RESULTS Seven out of 170 eligible studies met our inclusion criteria. A total of 207 advanced HNSCC patients were evaluated with F-FDG PET/CT at baseline and after IC in the selected articles. Six from seven studies concluded that F-FDG PET/CT allowed early evaluation response to IC and predicted survival outcomes. CONCLUSION The present systematic review confirms the potential value of F-FDG PET/CT as a diagnostic tool for early IV response assessment in HNSCC patients. However, the lack of standard definitions for response criteria and heterogeneous IC protocols indicate the need to further studies in order to better define the role of F-FDG PET/CT in these patients.
Collapse
|
14
|
Serkova NJ, Eckhardt SG. Metabolic Imaging to Assess Treatment Response to Cytotoxic and Cytostatic Agents. Front Oncol 2016; 6:152. [PMID: 27471678 PMCID: PMC4946377 DOI: 10.3389/fonc.2016.00152] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/07/2016] [Indexed: 12/24/2022] Open
Abstract
For several decades, cytotoxic chemotherapeutic agents were considered the basis of anticancer treatment for patients with metastatic tumors. A decrease in tumor burden, assessed by volumetric computed tomography and magnetic resonance imaging, according to the response evaluation criteria in solid tumors (RECIST), was considered as a radiological response to cytotoxic chemotherapies. In addition to RECIST-based dimensional measurements, a metabolic response to cytotoxic drugs can be assessed by positron emission tomography (PET) using (18)F-fluoro-thymidine (FLT) as a radioactive tracer for drug-disrupted DNA synthesis. The decreased (18)FLT-PET uptake is often seen concurrently with increased apparent diffusion coefficients by diffusion-weighted imaging due to chemotherapy-induced changes in tumor cellularity. Recently, the discovery of molecular origins of tumorogenesis led to the introduction of novel signal transduction inhibitors (STIs). STIs are targeted cytostatic agents; their effect is based on a specific biological inhibition with no immediate cell death. As such, tumor size is not anymore a sensitive end point for a treatment response to STIs; novel physiological imaging end points are desirable. For receptor tyrosine kinase inhibitors as well as modulators of the downstream signaling pathways, an almost immediate inhibition in glycolytic activity (the Warburg effect) and phospholipid turnover (the Kennedy pathway) has been seen by metabolic imaging in the first 24 h of treatment. The quantitative imaging end points by magnetic resonance spectroscopy and metabolic PET (including 18F-fluoro-deoxy-glucose, FDG, and total choline) provide an early treatment response to targeted STIs, before a reduction in tumor burden can be seen.
Collapse
Affiliation(s)
- Natalie J. Serkova
- Department of Anesthesiology, University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
- Developmental Therapeutics Program, University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
| | - S. Gail Eckhardt
- Developmental Therapeutics Program, University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
- Division of Medical Oncology, Anschutz Medical Center, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
15
|
Early 18F-FDG PET/CT Evaluation Shows Heterogeneous Metabolic Responses to Anti-EGFR Therapy in Patients with Metastatic Colorectal Cancer. PLoS One 2016; 11:e0155178. [PMID: 27196139 PMCID: PMC4873260 DOI: 10.1371/journal.pone.0155178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/23/2016] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE The aim of this pilot study was to explore intrapatient mixed metabolic response and early 18F-FDG PET response evaluation using predefined quantification strategies in patients with advanced KRAS wild-type colorectal adenocarcinoma (mCRC) treated with cetuximab. METHODS A 18F-FDG PET was performed at baseline and after 2 cycles of cetuximab. Metabolic response was categorized using thresholds suggested in PERCIST. Quantitative analysis was done for the sum of all target lesions, ≤ 5 lesions and the metabolically most active lesion per PET. Quantitative data were correlated with clinical benefit, according to RECIST v1.1, after two months of treatment. RESULTS In nine evaluable patients the total number of target lesions was 34 (1-8 per patient). Mixed metabolic response was observed in three out of seven patients with multiple target lesions, using TLG. Dichotomised metabolic data of the sum of all or ≤ 5 lesions had a concordance with clinical benefit of 89% using SULmax or SULpeak, and 100% using TLG. Evaluating the metabolically most active lesion, concordance was 89% for all three units. Additionally, the decrease in TLG was significantly correlated with PFS for all three quantification strategies. CONCLUSION Mixed metabolic response was observed in nearly half of the patients with advanced KRAS wild-type mCRC treated with cetuximab. If ≤ 5 target lesions were evaluated using TLG clinical benefit was predicted correctly for all patients. Moreover, decrease in TLG is significantly correlated with the duration of PFS. Validation of these promising preliminary results in a larger cohort is currently on-going. TRIAL REGISTRATION ClinicalTrials.gov NCT01691391.
Collapse
|
16
|
Schindler E, Amantea MA, Karlsson MO, Friberg LE. PK-PD modeling of individual lesion FDG-PET response to predict overall survival in patients with sunitinib-treated gastrointestinal stromal tumor. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:173-81. [PMID: 27299707 PMCID: PMC4846778 DOI: 10.1002/psp4.12057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022]
Abstract
Pharmacometric models were developed to characterize the relationships between lesion-level tumor metabolic activity, as assessed by the maximum standardized uptake value (SUVmax) obtained on [(18)F]-fluorodeoxyglucose (FDG) positron emission tomography (PET), tumor size, and overall survival (OS) in 66 patients with gastrointestinal stromal tumor (GIST) treated with intermittent sunitinib. An indirect response model in which sunitinib stimulates tumor loss best described the typically rapid decrease in SUVmax during on-treatment periods and the recovery during off-treatment periods. Substantial interindividual and interlesion variability were identified in SUVmax baseline and drug sensitivity. A parametric time-to-event model identified the relative change in SUVmax at one week for the lesion with the most pronounced response as a better predictor of OS than tumor size. Based on the proposed modeling framework, early changes in FDG-PET response may serve as predictor for long-term outcome in sunitinib-treated GIST.
Collapse
Affiliation(s)
- E Schindler
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - M O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Challapalli A, Aboagye EO. Positron Emission Tomography Imaging of Tumor Cell Metabolism and Application to Therapy Response Monitoring. Front Oncol 2016; 6:44. [PMID: 26973812 PMCID: PMC4770188 DOI: 10.3389/fonc.2016.00044] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/12/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer cells do reprogram their energy metabolism to enable several functions, such as generation of biomass including membrane biosynthesis, and overcoming bioenergetic and redox stress. In this article, we review both established and evolving radioprobes developed in association with positron emission tomography (PET) to detect tumor cell metabolism and effect of treatment. Measurement of enhanced tumor cell glycolysis using 2-deoxy-2-[(18)F]fluoro-D-glucose is well established in the clinic. Analogs of choline, including [(11)C]choline and various fluorinated derivatives are being tested in several cancer types clinically with PET. In addition to these, there is an evolving array of metabolic tracers for measuring intracellular transport of glutamine and other amino acids or for measuring glycogenesis, as well as probes used as surrogates for fatty acid synthesis or precursors for fatty acid oxidation. In addition to providing us with opportunities for examining the complex regulation of reprogramed energy metabolism in living subjects, the PET methods open up opportunities for monitoring pharmacological activity of new therapies that directly or indirectly inhibit tumor cell metabolism.
Collapse
Affiliation(s)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
18
|
Multimodal imaging with (18)F-FDG-PET/CT and (111)In-Octreotide SPECT in patients with metastatic medullary thyroid carcinoma. Ann Nucl Med 2016; 30:234-41. [PMID: 26753628 DOI: 10.1007/s12149-015-1056-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/13/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The aim of our study was to determine the role of fluorine-18 fluorodeoxyglucose positron emission tomography/computed tomography ((18)F-FDG-PET/CT) and indium-111 Octreotide single photon emission tomography ((111)In-Octreotide SPECT) in the evaluation of metastatic medullary thyroid carcinoma (MMTC). METHODS Twenty-five MMTC patients were retrospectively evaluated. All patients had undergone whole-body (18)F-FDG-PET/CT including 20 who had also undergone (111)In-Octreotide SPECT within a maximum interval of 6 weeks. Diagnostic contrast-enhanced computed tomography (CT) alone or as part of (18)F-FDG-PET/CT examination was performed in all patients. RESULTS Contrast-enhanced CT detected a total of 131 lesions including 79 enlarged lymph nodes and 14 bone lesions. (18)F-FDG-PET/CT visualized a total of 92 true positive lesions (SUVmax range 1.1-10.0, mean 4.0 ± 1.7) including 66 lymph nodes, 7 of which were not enlarged on CT, and 8 bone metastases. In the 20 patients studied with both techniques, a total of 64 and 46 true positive lesions were detected by (18)F-FDG-PET/CT and (111)In-Octreotide SPECT, respectively. In particular, (18)F-FDG uptake was found in 43 lymph nodes and in 7 bone metastases whereas (111)In-Octreotide uptake was detected in 27 lymph nodes and in 10 bone metastases. CONCLUSIONS In MMTC patients, (18)F-FDG-PET/CT provides a useful contribution mainly in evaluating lymph node involvement whereas (111)In-Octreotide SPECT can contribute to the detection and somatostatin receptor characterization especially of bone lesions.
Collapse
|
19
|
Heijmen L, Ter Voert EGW, Punt CJA, Heerschap A, Oyen WJG, Bussink J, Sweep CGJ, Laverman P, Span PN, de Geus-Oei LF, Boerman OC, van Laarhoven HWM. Monitoring hypoxia and vasculature during bevacizumab treatment in a murine colorectal cancer model. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 9:237-45. [PMID: 24700751 DOI: 10.1002/cmmi.1564] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 07/24/2013] [Accepted: 08/01/2013] [Indexed: 01/17/2023]
Abstract
The purpose of this study was to assess the effect of bevacizumab on vasculature and hypoxia in a colorectal tumor model. Nude mice with subcutaneous LS174T tumors were treated with bevacizumab or saline. To assess tumor properties, separate groups of mice were imaged using (18) F-Fluoromisonidazole (FMISO) and (18) F-Fluorodeoxyglucose (FDG) positron emission tomography or magnetic resonance imaging before and 2, 6 and 10 days after the start of treatment. Tumors were harvested after imaging to determine hypoxia and vascular density immunohistochemically. The T2 * time increased significantly less in the bevacizumab group. FMISO uptake increased more over time in the control group. Vessel density significantly decreased in the bevacizumab-treated group. The Carbonic anhydrase 9 (CAIX) and glucose uptake transporter 1 (GLUT1) fractions were higher in bevacizumab-treated tumors. However, the hypoxic fraction showed no significant difference. Bevacizumab led to shorter T2 * times and higher GLUT1 and CAIX expression, suggesting an increase in hypoxia and a higher glycolytic rate. This could be a mechanism of resistance to bevacizumab. The increase in hypoxia, however, could not be demonstrated by pimonidazole/FMISO, possibly because distribution of these tracers is hampered by bevacizumab-induced effects on vascular permeability and perfusion.
Collapse
Affiliation(s)
- L Heijmen
- Department of Medical Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lalami Y, Garcia C, Flamen P, Ameye L, Paesmans M, Awada A. Phase II trial evaluating the efficacy of sorafenib (BAY 43-9006) and correlating early fluorodeoxyglucose positron emission tomography-CT response to outcome in patients with recurrent and/or metastatic head and neck cancer. Head Neck 2015; 38:347-54. [DOI: 10.1002/hed.23898] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 07/11/2014] [Accepted: 10/16/2014] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yassine Lalami
- Department of Medical Oncology, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| | - Camillo Garcia
- Department of Nuclear Medicine, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| | - Lieveke Ameye
- Data Center, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| | - Marianne Paesmans
- Data Center, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| | - Ahmad Awada
- Department of Medical Oncology, Institut Jules Bordet; Université de Bruxelles; Brussels Belgium
| |
Collapse
|
21
|
Alam IS, Arshad MA, Nguyen QD, Aboagye EO. Radiopharmaceuticals as probes to characterize tumour tissue. Eur J Nucl Med Mol Imaging 2015; 42:537-61. [PMID: 25647074 DOI: 10.1007/s00259-014-2984-3] [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: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 01/06/2023]
Abstract
Tumour cells exhibit several properties that allow them to grow and divide. A number of these properties are detectable by nuclear imaging methods. We discuss crucial tumour properties that can be described by current radioprobe technologies, further discuss areas of emerging radioprobe development, and finally articulate need areas that our field should aspire to develop. The review focuses largely on positron emission tomography and draws upon the seminal 'Hallmarks of Cancer' review article by Hanahan and Weinberg in 2011 placing into context the present and future roles of radiotracer imaging in characterizing tumours.
Collapse
Affiliation(s)
- Israt S Alam
- Comprehensive Cancer Imaging Centre, Imperial College London, London, W12 0NN, UK
| | | | | | | |
Collapse
|
22
|
Multiparametric monitoring of early response to antiangiogenic therapy: a sequential perfusion CT and PET/CT study in a rabbit VX2 tumor model. ScientificWorldJournal 2014; 2014:701954. [PMID: 25383376 PMCID: PMC4213998 DOI: 10.1155/2014/701954] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/19/2014] [Accepted: 09/01/2014] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES To perform dual analysis of tumor perfusion and glucose metabolism using perfusion CT and FDG-PET/CT for the purpose of monitoring the early response to bevacizumab therapy in rabbit VX2 tumor models and to assess added value of FDG-PET to perfusion CT. METHODS Twenty-four VX2 carcinoma tumors implanted in bilateral back muscles of 12 rabbits were evaluated. Serial concurrent perfusion CT and FDG-PET/CT were performed before and 3, 7, and 14 days after bevacizumab therapy (treatment group) or saline infusion (control group). Perfusion CT was analyzed to calculate blood flow (BF), blood volume (BV), and permeability surface area product (PS); FDG-PET was analyzed to calculate SUVmax, SUVmean, total lesion glycolysis (TLG), entropy, and homogeneity. The flow-metabolic ratio (FMR) was also calculated and immunohistochemical analysis of microvessel density (MVD) was performed. RESULTS On day 14, BF and BV in the treatment group were significantly lower than in the control group. There were no significant differences in all FDG-PET-derived parameters between both groups. In the treatment group, FMR prominently decreased after therapy and was positively correlated with MVD. CONCLUSIONS In VX2 tumors, FMR could provide further insight into the early antiangiogenic effect reflecting a mismatch in intratumor blood flow and metabolism.
Collapse
|
23
|
Gallamini A, Zwarthoed C, Borra A. Positron Emission Tomography (PET) in Oncology. Cancers (Basel) 2014; 6:1821-89. [PMID: 25268160 PMCID: PMC4276948 DOI: 10.3390/cancers6041821] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/25/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
Since its introduction in the early nineties as a promising functional imaging technique in the management of neoplastic disorders, FDG-PET, and subsequently FDG-PET/CT, has become a cornerstone in several oncologic procedures such as tumor staging and restaging, treatment efficacy assessment during or after treatment end and radiotherapy planning. Moreover, the continuous technological progress of image generation and the introduction of sophisticated software to use PET scan as a biomarker paved the way to calculate new prognostic markers such as the metabolic tumor volume (MTV) and the total amount of tumor glycolysis (TLG). FDG-PET/CT proved more sensitive than contrast-enhanced CT scan in staging of several type of lymphoma or in detecting widespread tumor dissemination in several solid cancers, such as breast, lung, colon, ovary and head and neck carcinoma. As a consequence the stage of patients was upgraded, with a change of treatment in 10%-15% of them. One of the most evident advantages of FDG-PET was its ability to detect, very early during treatment, significant changes in glucose metabolism or even complete shutoff of the neoplastic cell metabolism as a surrogate of tumor chemosensitivity assessment. This could enable clinicians to detect much earlier the effectiveness of a given antineoplastic treatment, as compared to the traditional radiological detection of tumor shrinkage, which usually takes time and occurs much later.
Collapse
Affiliation(s)
- Andrea Gallamini
- Department of Research and Medical Innovation, Antoine Lacassagne Cancer Center, Nice University, Nice Cedex 2-06189 Nice, France.
| | - Colette Zwarthoed
- Department of Nuclear Medicine, Antoine Lacassagne Cancer Center, Nice University, Nice Cedex 2-06189 Nice, France.
| | - Anna Borra
- Hematology Department S. Croce Hospital, Via M. Coppino 26, Cuneo 12100, Italy.
| |
Collapse
|
24
|
Skougaard K, Johannesen HH, Nielsen D, Schou JV, Jensen BV, Høgdall EVS, Hendel HW. CT versus FDG-PET/CT response evaluation in patients with metastatic colorectal cancer treated with irinotecan and cetuximab. Cancer Med 2014; 3:1294-301. [PMID: 24941936 PMCID: PMC4302679 DOI: 10.1002/cam4.271] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 04/21/2014] [Accepted: 04/30/2014] [Indexed: 12/26/2022] Open
Abstract
We compared morphologic computed tomography (CT)-based to metabolic fluoro-deoxy-glucose (FDG) positron emission tomography (PET)/CT-based response evaluation in patients with metastatic colorectal cancer and correlated the findings with survival and KRAS status. From 2006 to 2009, patients were included in a phase II trial and treated with cetuximab and irinotecan every second week. They underwent FDG-PET/CT examination at baseline and after every fourth treatment cycle. Response evaluation was performed prospectively according to Response Evaluation Criteria in Solid Tumors (RECIST 1.0) and retrospectively according to Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST). Best overall responses were registered. Sixty-one patients were eligible for response evaluation. Partial response (PR) rate was 18%, stable disease (SD) rate 64%, and progressive disease (PD) rate 18%. Partial metabolic response (PMR) rate was 56%, stable metabolic disease rate 33%, and progressive metabolic disease (PMD) rate 11%. Response agreement was poor, κ-coefficient 0.19. Hazard ratio for overall survival for responders (PR/PMR) versus nonresponders (PD/PMD) was higher for CT- than for FDG-PET/CT evaluation. Within patients with KRAS mutations, none had PR but 44% had PMR. In conclusion, morphologic and metabolic response agreement was poor primarily because a large part of the patients shifted from SD with CT evaluation to PMR when evaluated with FDG-PET/CT. Furthermore, a larger fraction of the patients with KRAS mutations had a metabolic treatment response.
Collapse
Affiliation(s)
- Kristin Skougaard
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | | | | | | | | | | | |
Collapse
|
25
|
Schelhaas S, Wachsmuth L, Viel T, Honess DJ, Heinzmann K, Smith DM, Hermann S, Wagner S, Kuhlmann MT, Müller-Tidow C, Kopka K, Schober O, Schäfers M, Schneider R, Aboagye EO, Griffiths J, Faber C, Jacobs AH. Variability of Proliferation and Diffusion in Different Lung Cancer Models as Measured by 3'-Deoxy-3'-¹⁸F-Fluorothymidine PET and Diffusion-Weighted MR Imaging. J Nucl Med 2014; 55:983-8. [PMID: 24777288 DOI: 10.2967/jnumed.113.133348] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/15/2014] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Molecular imaging allows the noninvasive assessment of cancer progression and response to therapy. The aim of this study was to investigate molecular and cellular determinants of 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) PET and diffusion-weighted (DW) MR imaging in lung carcinoma xenografts. METHODS Four lung cancer cell lines (A549, HTB56, EBC1, and H1975) were subcutaneously implanted in nude mice, and growth was followed by caliper measurements. Glucose uptake and tumor proliferation were determined by (18)F-FDG and (18)F-FLT PET, respectively. T2-weighted MR imaging was performed, and the apparent diffusion coefficient (ADC) was determined by DW MR imaging as an indicator of cell death. Imaging findings were correlated to histology with markers for tumor proliferation (Ki67, 5-bromo-2'-deoxyuridine [BrdU]) and cell death (caspase-3, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling). The expression of human equilibrative nucleoside transporter 1 (hENT1), thymidine kinase 1 (TK1), thymidylate synthase, and thymidine phosphorylase (TP) were analyzed by Western blot and immunohistochemistry. Thymidine levels were determined by liquid chromatography-mass spectrometry. RESULTS Xenografts varied with respect to in vivo growth rates. MR imaging and PET revealed intratumoral heterogeneities, which were confirmed by histology. (18)F-FLT uptake differed significantly between tumor lines, with A549 and H1975 demonstrating the highest radiotracer accumulation (A549, 8.5 ± 3.2; HTB56, 4.4 ± 0.7; EBC1, 4.4 ± 1.2; and H1975, 12.1 ± 3.5 maximal percentage injected dose per milliliter). In contrast, differences in (18)F-FDG uptake were only marginal. No clear relationship between (18)F-FLT accumulation and immunohistochemical markers for tumor proliferation (Ki67, BrdU) as well as hENT1, TK1, or TS expression was detected. However, TP was highly expressed in A549 and H1975 xenografts, which was accompanied by low tumor thymidine concentrations, suggesting that tumor thymidine levels influence (18)F-FLT uptake in the tumor models investigated. MR imaging revealed higher ADC values within proliferative regions of H1975 and A549 tumors than in HTB56 and EBC1. These ADC values were negatively correlated with cell density but not directly related to cell death. CONCLUSION A direct relationship of (18)F-FLT with proliferation or ADC with cell death might be complicated by the interplay of multiple processes at the cellular and physiologic levels in untreated tumors. This issue must be considered when using these imaging modalities in preclinical or clinical settings.
Collapse
Affiliation(s)
- Sonja Schelhaas
- 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
| | - Thomas Viel
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Davina J Honess
- Cancer Research United Kingdom Cambridge Institute, Cambridge, United Kingdom
| | - Kathrin Heinzmann
- Cancer Research United Kingdom Cambridge Institute, Cambridge, United Kingdom
| | | | - Sven Hermann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Stefan Wagner
- Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Michael T Kuhlmann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | - Carsten Müller-Tidow
- Department of Hematology and Oncology, University Hospital of Münster, Münster, Germany
| | - Klaus Kopka
- Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Otmar Schober
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany Department of Nuclear Medicine, University Hospital of Münster, Münster, Germany
| | | | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, London, United Kingdom; and
| | - John Griffiths
- Cancer Research United Kingdom Cambridge Institute, 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
|
Role of ¹⁸F-FDG PET CT as an independent prognostic indicator in patients with hepatocellular carcinoma. Nucl Med Commun 2014; 34:749-57. [PMID: 23689586 DOI: 10.1097/mnm.0b013e3283622eef] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate the role of F-fluorodeoxyglucose PET computed tomography (F-FDG PET CT) as an independent prognostic indicator in patients with hepatocellular carcinoma (HCC). MATERIALS AND METHODS PET contrast-enhanced CT scans of 100 consecutive patients with HCC were reviewed retrospectively. Patients were asked to fast for 6 h before the study and blood glucose levels were monitored and ensured to be less than 200 mg/dl before injection of F-FDG. After administering the F-FDG injection (370-550 MBq) patients were instructed to rest comfortably for 45-60 min. All images were acquired using a dedicated GE Discovery PET/CT scanner. The PET CT scans of all the patients were reported separately by two nuclear medicine physicians. A stage-wise analysis of the compiled data was carried out. Lesions that showed standardized uptake values greater than background activity (activity in adjacent normal liver tissue) were defined as having increased F-FDG uptake. Pearson's χ -test or the Kruskal-Wallis test was used to assess statistical significance. A P value less than 0.05 was taken as significant. RESULTS In this retrospective study of 100 HCC patients, a radiologically higher-stage disease was found more commonly in patients with F-FDG-avid primary tumors (P<0.001), whereas a lower-stage disease was found in patients with non-F-FDG-avid primary tumors. The non-F-FDG-avid tumors also showed lower incidence of metastatic disease and portal vein thrombosis (P<0.001). The histopathological findings of the patients who underwent liver transplantation demonstrated that a higher-grade tumor was more common in the F-FDG-avid tumor group than in the non-F-FDG-avid tumor group (P<0.05). CONCLUSION An F-FDG PET CT scan can be used not only for staging but also as a tool for preoperative prediction of cellular differentiation in patients with HCC. The F-FDG uptake seen on a PET scan can serve as a molecular signature for management decisions and can be used as an independent and significant prognostic factor in patients with HCC.
Collapse
|
27
|
Merchant S, Witney TH, Aboagye EO. Imaging as a pharmacodynamic and response biomarker in cancer. Clin Transl Imaging 2014. [DOI: 10.1007/s40336-014-0049-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
2-Deoxy-2-[18F]fluoro-D-glucose positron emission tomography demonstrates target inhibition with the potential to predict anti-tumour activity following treatment with the AKT inhibitor AZD5363. Mol Imaging Biol 2014; 15:476-85. [PMID: 23344784 DOI: 10.1007/s11307-013-0613-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE The phosphatidyl inositol 3 kinase, AKT and mammalian target of rapamycin are frequently deregulated in human cancer and are among one of the most promising targets for cancer therapy. AZD5363 (AstraZeneca) is an AKT inhibitor in phase 1 clinical trials. Given its utility in assessing glucose metabolism, we investigated the role of 2-Deoxy-2-[18F]fluoro-D-glucose (18F-FDG) positron emission tomography (PET) as a biomarker to demonstrate target inhibition and its potential to predict and demonstrate the anti-tumour activity of AZD5363. METHODS 18F-FDG PETscans were performed in nude mice in a number of xenograft models (U87-MG glioblastoma, BT474C breast carcinoma and Calu-6 lung). Mice were fasted prior to imaging, and either static or dynamic 18F-FDG PET imaging was performed. RESULTS We have shown that 18F-FDG uptake in tumour xenografts was reduced by 39% reduction compared to vehicle after a single dose of AZD5363, demonstrating activation of the AKT pathway after only 4 h of dosing. Multiple doses of AZD5363 showed an anti-tumour volume effect and a reduction in 18F-FDG uptake (28% reduction compared to vehicle), highlighting the potential of 18F-FDG PET as an efficacy biomarker. Furthermore, the degree of inhibition of 18F-FDG uptake corresponded with the sensitivity of the tumour model to AZD5363. The use of dynamic 18F-FDG PET and a two-compartmental analysis identified the mechanism of this change to be due to a change in cellular uptake of 18F-FDG following administration of AZD5363. CONCLUSIONS We conclude that 18F-FDG PET is a promising pharmacodynamic biomarker of AKT pathway inhibition, with potential to predict and demonstrate anti-tumour activity. It is a biomarker that may stop ineffective drug schedules, helping to make early stop decisions and identify responding subsets of patients, resulting in improved clinical decision making both during drug development and patient management.
Collapse
|
29
|
Abstract
Over recent years, there has been a rapid expansion in our knowledge of the factors that regulate tumor growth; this has resulted in the identification of new therapeutic targets and improvements in the long-term survival of cancer patients. New noninvasive biomarkers of drug targets and pathway modulation in vivo are needed to guide therapy selection and detect drug resistance early so that alternative, more effective treatments can be offered. The translation of new therapeutics into the clinic is disappointingly slow, expensive, and subject to high rates of attrition often occurring at late stages (phase 3) of development. In an attempt to mitigate these delays and failures, there has been resurgence in the development of new molecular imaging probes for studies with positron emission tomography (PET) to characterize tumor biology. In the assessment of therapeutic effects, PET allows imaging of entire tumor burden in a noninvasive repeatable manner. This chapter focuses on the clinical translation of PET imaging agents from bench to bedside. New probes are being used to study a diverse range of processes such as angiogenesis, apoptosis, fatty acid metabolism, and growth factor receptor expression. In the future, validation of these novel imaging probes could allow more innovative therapies to be adapted earlier in the clinic leading to improved patient outcomes.
Collapse
Affiliation(s)
- Laura M Kenny
- Comprehensive Cancer Imaging Centre, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Department of Surgery & Cancer, Imperial College London, London, United Kingdom.
| |
Collapse
|
30
|
Abstract
Somatic evolution during cancer progression and therapy results in tumour cells that show a wide range of phenotypes, which include rapid proliferation and quiescence. Evolutionary life history theory may help us to understand the diversity of these phenotypes. Fast life history organisms reproduce rapidly, whereas those with slow life histories show less fecundity and invest more resources in survival. Life history theory also provides an evolutionary framework for phenotypic plasticity, which has potential implications for understanding 'cancer stem cells'. Life history theory suggests that different therapy dosing schedules might select for fast or slow life history cell phenotypes, with important clinical consequences.
Collapse
Affiliation(s)
- C Athena Aktipis
- 1] Center for Evolution and Cancer and the Department of Surgery, University of California San Francisco, 2340 Sutter Street, BOX 1351, San Francisco, California 94143-1351, USA. [2] Department of Psychology, Arizona State University, PO Box 871104, Tempe, Arizona 85287-1104, USA
| | | | | | | | | |
Collapse
|
31
|
Jung KH, Lee JH, Thien Quach CH, Paik JY, Oh H, Park JW, Lee EJ, Moon SH, Lee KH. Resveratrol Suppresses Cancer Cell Glucose Uptake by Targeting Reactive Oxygen Species–Mediated Hypoxia-Inducible Factor-1α Activation. J Nucl Med 2013; 54:2161-7. [DOI: 10.2967/jnumed.112.115436] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
32
|
Bendell JC, Gordon MS, Hurwitz HI, Jones SF, Mendelson DS, Blobe GC, Agarwal N, Condon CH, Wilson D, Pearsall AE, Yang Y, McClure T, Attie KM, Sherman ML, Sharma S. Safety, pharmacokinetics, pharmacodynamics, and antitumor activity of dalantercept, an activin receptor-like kinase-1 ligand trap, in patients with advanced cancer. Clin Cancer Res 2013; 20:480-9. [PMID: 24173543 DOI: 10.1158/1078-0432.ccr-13-1840] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The angiogenesis inhibitor dalantercept (formerly ACE-041) is a soluble form of activin receptor-like kinase-1 (ALK1) that prevents activation of endogenous ALK1 by bone morphogenetic protein-9 (BMP9) and BMP10 and exhibits antitumor activity in preclinical models. This first-in-human study of dalantercept evaluated its safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity in adults with advanced solid tumors. EXPERIMENTAL DESIGN Patients in dose-escalating cohorts received dalantercept subcutaneously at one of seven dose levels (0.1-4.8 mg/kg) every 3 weeks until disease progression. Patients in an expansion cohort received dalantercept at 0.8 or 1.6 mg/kg every 3 weeks until disease progression. RESULTS In 37 patients receiving dalantercept, the most common treatment-related adverse events were peripheral edema, fatigue, and anemia. Edema and fluid retention were dose-limiting toxicities and responded to diuretic therapy. No clinically significant, treatment-related hypertension, proteinuria, gross hemorrhage, or gastrointestinal perforations were observed. One patient with refractory squamous cell cancer of the head and neck had a partial response, and 13 patients had stable disease according to RECISTv1.1, eight of whom had prolonged periods (≥12 weeks) of stable disease. Correlative pharmacodynamic markers included tumor metabolic activity and tumor blood flow, which decreased from baseline in 63% and 82% of evaluable patients, respectively, and telangiectasia in eight patients. CONCLUSION Dalantercept was well-tolerated at doses up to 1.6 mg/kg, with a safety profile distinct from inhibitors of the VEGF pathway. Dalantercept displayed promising antitumor activity in patients with advanced refractory cancer, and multiple phase II studies are underway.
Collapse
Affiliation(s)
- Johanna C Bendell
- Authors' Affiliations: Sarah Cannon Research Institute, Nashville, Tennessee; Pinnacle Oncology Hematology, Scottsdale, Arizona; Duke University Medical Center, Durham, North Carolina; Huntsman Cancer Institute, Salt Lake City, Utah; and Acceleron Pharma, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
(18)F-FDG Is a Surrogate Marker of Therapy Response and Tumor Recovery after Drug Withdrawal during Treatment with a Dual PI3K/mTOR Inhibitor in a Preclinical Model of Cisplatin-Resistant Ovarian Cancer. Transl Oncol 2013; 6:586-95. [PMID: 24151539 DOI: 10.1593/tlo.13100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 07/21/2013] [Accepted: 07/24/2013] [Indexed: 01/09/2023] Open
Abstract
AIM Targeting the phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway is a potential means of overcoming chemoresistance in ovarian cancer. We investigated the capability of (18)F-fluororodeoxyglucose ((18)F-FDG) small-animal positron emission tomography (SA-PET) to predict the effects of a dual PI3K/mTOR inhibitor (BEZ-235) in a cisplatin-resistant ovarian cancer model. METHODS In a first experiment, nude rats bearing subcutaneous SKOV3 tumors received BEZ-235 for 3 days given alone or after paclitaxel and were compared to controls (either untreated or that were given the excipients of paclitaxel and BEZ-235). SA-PET was performed at baseline, on day 3, and day 7. In a second experiment aiming at further exploring the kinetics of (18)F-FDG tumor uptake during the first 48 hours following drug cessation, untreated controls were compared to rats receiving BEZ-235, which were imaged at baseline, on day 3, on day 4, and on day 5. SA-PET results were compared to cell proliferation assessment (Ki-67), PI3K/mTOR downstream target expression studies (pAKT and phospho-eukaryotic translation initiation factor 4E-binding protein 1), and apoptosis evaluation (cleaved caspase-3). RESULTS In the first experiment, BEZ-235, compared to untreated controls, induced a marked decrease in (18)F-FDG uptake on day 3, which was correlated to a significant decrease in cell proliferation and to a significant PI3K/mTOR pathway inhibition. No tumor necrosis or apoptosis occurred. Four days following treatment cessation, tumor recovery (in terms of PI3K/mTOR inhibition and cell proliferation) occurred and was identified by (18)F-FDG SA-PET. Paclitaxel plus BEZ-235 showed results similar to BEZ-235 alone. In the second experiment, PI3K/mTOR pathways exhibited partial recovery as early as 24 hours following treatment cessation, but both (18)F-FDG SA-PET and cell proliferation remained unchanged. CONCLUSIONS (18)F-FDG SA-PET is a surrogate marker of target inhibition during treatment with BEZ-235 and predicts tumor recovery 4 days after drug withdrawal, but not during the first 48 hours following drug cessation, when a lag between PI3K/mTOR pathway recovery and metabolic recovery is observed. (18)F-FDG SA-PET could be used for therapy monitoring of PI3K/mTOR inhibitors, but our results also raise questions regarding the potential impact of the delay between PET imaging and the last drug intake on the accuracy of FDG imaging.
Collapse
|
34
|
A radiologist's guide to treatment response criteria in oncologic imaging: anatomic imaging biomarkers. AJR Am J Roentgenol 2013; 201:237-45. [PMID: 23883205 DOI: 10.2214/ajr.12.9862] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The purpose of this article is to describe the imaging biomarkers of treatment response and provide an overview of anatomic imaging biomarkers. CONCLUSION Imaging biomarkers of treatment response have evolved into the primary endpoint of response in most phase 2 studies. Anatomic imaging biomarkers are applied to depict change in tumor size in response to treatment and are currently the most commonly applied method of treatment response evaluation.
Collapse
|
35
|
A radiologist's guide to treatment response criteria in oncologic imaging: functional, molecular, and disease-specific imaging biomarkers. AJR Am J Roentgenol 2013; 201:246-56. [PMID: 23883206 DOI: 10.2214/ajr.12.9878] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This article reviews the functional, molecular, and disease-specific imaging biomarkers of treatment response. CONCLUSION Substantial progress has been made in the evolution of drugs directed at specific targets of the tumor lifecycle. These novel agents are predominantly cytostatic, and their efficacy may be optimally evaluated by functional, molecular, and disease-specific imaging biomarkers.
Collapse
|
36
|
Gillebert Q, Gligorov J, Kerrou K. Modification du métabolisme tumoral au cours du traitement : suivi de la réponse thérapeutique par tomographie par émission de positons. ONCOLOGIE 2013. [DOI: 10.1007/s10269-013-2322-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Tirumani SH, Jagannathan JP, O'Regan K, Kim KW, Shinagare AB, Krajewski KM, Ramaiya NH. Molecular targeted therapies in non-GIST soft tissue sarcomas: what the radiologist needs to know. Cancer Imaging 2013; 13:197-211. [PMID: 23649384 PMCID: PMC3645342 DOI: 10.1102/1470-7330.2013.0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2013] [Indexed: 12/30/2022] Open
Abstract
Non-gastrointestinal stromal soft tissue sarcomas are uncommon neoplasms that have a dismal prognosis due to a high incidence of metastases and a poor response to conventional chemotherapy. The identification of characteristic genetic alterations in several of these tumors has opened the window for molecular targeted therapies in patients who have failed conventional chemotherapy. Imaging plays a critical role in assessing the response to these novel therapeutic agents. Just like the response of gastrointestinal stromal tumors to imatinib, the response of non-gastrointestinal stromal soft tissue sarcomas to molecular targeted drugs is better evaluated on imaging by alternate tumor response criteria such as the Choi criteria. In addition, these drugs are associated with distinct class-specific drug toxicities that can come to attention for the first time on imaging. The purpose of this article is to provide a primer for the radiologist on the various molecular targeted therapies in advanced/metastatic non-gastrointestinal stromal soft tissue sarcomas with emphasis on the role of imaging in assessing treatment response and complications.
Collapse
Affiliation(s)
- Sree Harsha Tirumani
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Perumal M, Stronach EA, Gabra H, Aboagye EO. Evaluation of 2-deoxy-2-[18F]fluoro-D-glucose- and 3'-deoxy-3'-[18F]fluorothymidine-positron emission tomography as biomarkers of therapy response in platinum-resistant ovarian cancer. Mol Imaging Biol 2013; 14:753-61. [PMID: 22484552 DOI: 10.1007/s11307-012-0554-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE We evaluated whether 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG) and 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) positron emission tomography (PET) could be used as imaging biomarkers of platinum resensitization in ovarian cancer. PROCEDURES Paired platinum-sensitive and platinum-resistant ovarian cancer cells from the same patient, PEO1 and PEO4, grown as tumor xenografts in nude mice, were assessed by PET. RESULTS The AKT inhibitor, API-2, resensitized platinum-resistant PEO4 tumors to cisplatin, leading to a markedly lower Ki67 labeling index (p ≤ 0.006, n = 6 per group). [(18)F]FDG-PET and [(18)F]FLT-PET imaging variables were lower after combination treatment compared with vehicle treatment (p ≤ 0.006, n = 6 per group). No changes were seen with either drug alone. PRAS40 phosphorylation status was a sensitive biochemical marker of pathway inhibition, whereas reductions thymidine kinase 1 expression defined the [(18)F]FLT response. CONCLUSIONS Therapeutic inhibition of AKT activation in acquired platinum-resistant disease can be imaged noninvasively by [(18)F]FDG-PET and [(18)F]FLT-PET warranting further assessment.
Collapse
Affiliation(s)
- Meg Perumal
- Comprehensive Cancer Imaging Centre, Department of Cancer & Surgery, Faculty of Medicine, Imperial College London, Hammersmith Hospital, 240 MRC Cyclotron Building, Du Cane Road, London, W12 0NN, UK
| | | | | | | |
Collapse
|
39
|
Skougaard K, Nielsen D, Jensen BV, Hendel HW. Comparison of EORTC criteria and PERCIST for PET/CT response evaluation of patients with metastatic colorectal cancer treated with irinotecan and cetuximab. J Nucl Med 2013; 54:1026-31. [PMID: 23572497 DOI: 10.2967/jnumed.112.111757] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The study aim was to compare European Organization for Research and Treatment of Cancer (EORTC) criteria with PET Response Criteria in Solid Tumors (PERCIST) for response evaluation of patients with metastatic colorectal cancer treated with a combination of the chemotherapeutic drug irinotecan and the monoclonal antibody cetuximab. METHODS From 2006 to 2009, patients with metastatic colorectal cancer were prospectively included in a phase II trial evaluating the combination of irinotecan and cetuximab every second week, as third-line treatment. (18)F-FDG PET/CT was performed between 1 and 14 d before the first treatment and after every fourth treatment cycle until progression was identified by CT with Response Evaluation Criteria in Solid Tumors (RECIST). Response evaluation with (18)F-FDG PET/CT was retrospectively performed according to both EORTC criteria and PERCIST, classifying the patients into 4 response categories: complete metabolic response (CMR), partial metabolic response (PMR), stable metabolic disease (SMD), and progressive metabolic disease (PMD). Individual best overall metabolic response (BOmR) was registered with both sets of criteria, as well as survival within response categories, and compared. RESULTS A total of 61 patients and 203 PET/CT scans were eligible for response evaluation. With EORTC criteria, 38 had PMR, 16 had SMD, and 7 had PMD as their BOmR. With PERCIST, 34 had PMR, 20 had SMD, and 7 had PMD as their BOmR. None of the patients had CMR. There was agreement between EORTC criteria and PERCIST in 87% of the patients, and the corresponding κ-coefficient was 0.76. Disagreements were confined to PMR and SMD. Median overall survival (OS) in months with EORTC criteria was 14.2 in the PMR group and 7.2 in the combined SMD + PMD group. With PERCIST, it was 14.5 in the PMR group and 7.9 in the SMD + PMD group. CONCLUSION Response evaluation with EORTC criteria and PERCIST gave similar responses and OS outcomes with good agreement on BOmR (κ-coefficient, 0.76) and similar significant differences in median OS between response groups. Compared with EORTC criteria, we find PERCIST unambiguous because of clear definitions and therefore more straightforward to use.
Collapse
Affiliation(s)
- Kristin Skougaard
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark.
| | | | | | | |
Collapse
|
40
|
Sano K, Mitsunaga M, Nakajima T, Choyke PL, Kobayashi H. Acute cytotoxic effects of photoimmunotherapy assessed by 18F-FDG PET. J Nucl Med 2013; 54:770-5. [PMID: 23536226 DOI: 10.2967/jnumed.112.112110] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED We have recently developed a cancer-specific therapy, photoimmunotherapy, which uses an antibody-IR700 (phototoxic phthalocyanine dye) conjugate to bind to the cell membrane and near-infrared light to induce immediate and highly specific tumor killing in vivo. For monitoring the acute cytotoxic effects of photoimmunotherapy before the tumor begins to shrink, we used (18)F-FDG PET before and after this intervention in mice. METHODS Photoimmunotherapy was performed by binding panitumumab (anti-HER1)-IR700 to HER1-positive tumor cells (A431), followed by near-infrared light irradiation in vitro and in vivo. The uptake of (18)F-FDG in the tumor after photoimmunotherapy was evaluated in cellular uptake studies and PET imaging studies. Serial histologic analyses were conducted after photoimmunotherapy. RESULTS The in vitro cellular uptake of (18)F-FDG was reduced as the dose of light increased, and at high light dose (2 J/cm(2)) the uptake was reduced by more than 99% within 1 h after photoimmunotherapy. In vivo (18)F-FDG PET imaging showed that the accumulation of radioactivity in the treated tumors decreased 76% at 75 min after photoimmunotherapy and did not change for 24 h. In contrast, no significant changes were demonstrated in nontreated tumors. None of tumors changed size within 24 h after photoimmunotherapy, although diffuse necrosis was observed in photoimmunotherapy-treated tumors. CONCLUSION Immediate cytotoxic effects induced by photoimmunotherapy were clearly detected by decreased glucose uptake using (18)F-FDG PET even before changes in tumor size became evident. (18)F-FDG allows the clinical assessment of the therapeutic effects of photoimmunotherapy earlier than anatomic methods that rely on tumor size.
Collapse
Affiliation(s)
- Kohei Sano
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1088, USA
| | | | | | | | | |
Collapse
|
41
|
Evans MJ. Measuring oncogenic signaling pathways in cancer with PET: an emerging paradigm from studies in castration-resistant prostate cancer. Cancer Discov 2012; 2:985-94. [PMID: 23043150 PMCID: PMC3508522 DOI: 10.1158/2159-8290.cd-12-0178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
UNLABELLED As parallel advances in cancer biology and drug development continue to elevate the role of targeted therapies in oncology, the need for imaging biomarkers that systematically measure the biology associated with therapeutic intervention has become more urgent. Although the molecular imaging community has a commitment to develop technologies to this end, few investigational radiotracers directly measure the biology of common oncogenic signaling pathways often addressed by targeted therapies. Visible progress has been achieved with a handful of radiotracers rationally designed to intercalate the pathobiology of prostate cancer, a molecularly heterogeneous disease nevertheless broadly defined by a fairly small repertoire of recurrent oncogenic lesions. SIGNIFICANCE That variable treatment responses or emergent resistance phenotypes are often documented in humans argues strongly for diagnostic technologies that can be realistically applied posttherapy to capture the dynamic patterns of disease response. The purpose of this review is to describe a collection of radiotracers developed to measure the pathobiology of prostate cancer for improved treatment monitoring, placing particular emphasis on the biologic rationale for their preparation. A chronologic description of radiotracer development programs is outlined, primarily to stress how an ongoing dialectic between earlier and more contemporary imaging technologies has accelerated discovery.
Collapse
Affiliation(s)
- Michael J Evans
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
42
|
Figueiras RG, Padhani AR, Goh VJ, Vilanova JC, González SB, Martín CV, Caamaño AG, Naveira AB, Choyke PL. Novel oncologic drugs: what they do and how they affect images. Radiographics 2012; 31:2059-91. [PMID: 22084189 DOI: 10.1148/rg.317115108] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Targeted therapies are designed to interfere with specific aberrant biologic pathways involved in tumor development. The main classes of novel oncologic drugs include antiangiogenic drugs, antivascular agents, drugs interfering with EGFR-HER2 or KIT receptors, inhibitors of the PI3K/Akt/mTOR pathway, and hormonal therapies. Cancer cells usurp normal signal transduction pathways used by growth factors to stimulate proliferation and sustain viability. The interaction of growth factors with their receptors activates different intracellular pathways affecting key tumor biologic processes such as neoangiogenesis, tumor metabolism, and tumor proliferation. The response of tumors to anticancer therapy can be evaluated with anatomic response assessment, qualitative response assessment, and response assessment with functional and molecular imaging. Angiogenesis can be measured by means of perfusion imaging with computed tomography and magnetic resonance (MR) imaging. Diffusion-weighted MR imaging allows imaging evaluation of tumor cellularity. The main imaging techniques for studying tumor metabolism in vivo are positron emission tomography and MR spectroscopy. Familiarity with imaging findings secondary to tumor response to targeted therapies may help the radiologist better assist the clinician in accurate evaluation of tumor response to these anticancer treatments. Functional and molecular imaging techniques may provide valuable data and augment conventional assessment of tumor response to targeted therapies. Supplemental material available at http://radiographics.rsna.org/lookup/suppl/doi:10.1148/rg.317115108/-/DC1.
Collapse
Affiliation(s)
- Roberto García Figueiras
- Department of Radiology, Grupo de Imagen Molecular, Fundación IDICHUS/IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, Choupana s/n, 15702 Santiago de Compostela, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bengtsson T, Hicks RJ, Peterson A, Port RE. 18F-FDG PET as a surrogate biomarker in non-small cell lung cancer treated with erlotinib: newly identified lesions are more informative than standardized uptake value. J Nucl Med 2012; 53:530-7. [PMID: 22414632 DOI: 10.2967/jnumed.111.092544] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED This study assesses the predictive value of (18)F-FDG PET for overall survival in lung cancer patients treated with a targeted drug. METHODS (18)F-FDG PET was performed in 125 second- or third-line non-small cell lung cancer (NSCLC) patients with a baseline Eastern Cooperative Oncology Group performance status less than 3 before treatment with erlotinib (150 mg daily) and 2 wk into treatment. The predictive value of (18)F-FDG PET, clinical parameters, and epithelial growth factor receptor (EGFR) mutation status for survival duration was evaluated by fitting accelerated failure time models. RESULTS New lesions on PET at 2 wk, EGFR mutation status, performance status, and baseline tumor burden were independent and significant predictors of overall survival. Reduction of maximum standardized uptake value by at least 35% was predictive of survival only when EGFR mutation status was not accounted for. CONCLUSION (18)F-FDG PET in second- or third-line NSCLC patients at 2 wk after starting treatment with erlotinib carries information about overall survival. Parametric survival modeling enables a quantitative assessment of the predictive value of (18)F-FDG PET in the context of clinical and laboratory information. New-lesion status by (18)F-FDG PET at 2 wk is a potential surrogate biomarker for survival in NSCLC.
Collapse
|
44
|
Sharma R, Aboagye E. Development of radiotracers for oncology--the interface with pharmacology. Br J Pharmacol 2012; 163:1565-85. [PMID: 21175573 DOI: 10.1111/j.1476-5381.2010.01160.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is an increasing role for positron emission tomography (PET) in oncology, particularly as a component of early phase clinical trials. As a non-invasive functional imaging modality, PET can be used to assess both pharmacokinetics and pharmacodynamics of novel therapeutics by utilizing radiolabelled compounds. These studies can provide crucial information early in the drug development process that may influence the further development of novel therapeutics. PET imaging probes can also be used as early biomarkers of clinical response and to predict clinical outcome prior to the administration of therapeutic agents. We discuss the role of PET imaging particularly as applied to phase 0 studies and discuss the regulations involved in the development and synthesis of novel radioligands. The review also discusses currently available tracers and their role in the assessment of pharmacokinetics and pharmacodynamics as applied to oncology.
Collapse
Affiliation(s)
- Rohini Sharma
- Comprehensive Cancer Imaging Centre, Imperial College London Hammersmith Campus, Du Cane Road, London, UK
| | | |
Collapse
|
45
|
Nguyen QD, Challapalli A, Smith G, Fortt R, Aboagye EO. Imaging apoptosis with positron emission tomography: 'bench to bedside' development of the caspase-3/7 specific radiotracer [(18)F]ICMT-11. Eur J Cancer 2012; 48:432-40. [PMID: 22226480 DOI: 10.1016/j.ejca.2011.11.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 11/21/2011] [Indexed: 12/20/2022]
Abstract
The capacity to evade apoptosis has been defined as one of the hallmarks of cancer and, thus, effective anti-cancer therapy often induces apoptosis. A biomarker for imaging apoptosis could assist in monitoring the efficacy of a wide range of current and future therapeutics. Despite the potential, there are limited clinical examples of the use of positron emission tomography for imaging of apoptosis. [(18)F]ICMT-11 is a novel reagent designed to non-invasively image caspase-3 activation and, hence, drug-induced apoptosis. Radiochemistry development of [(18)F]ICMT-11 has been undertaken to improve specific radioactivity, reduce content of stable impurities, reduce synthesis time and enable automation for manufacture of multi-patient dose. Due to the promising mechanistic and safety profile of [(18)F]ICMT-11, the radiotracer is transitioning to clinical development and has been selected as a candidate radiotracer by the QuIC-ConCePT consortium for further evaluation in preclinical models and humans. A successful outcome will allow use of the radiotracer as qualified method for evaluating the pharmaceutical industry's next generation therapeutics.
Collapse
Affiliation(s)
- Quang-Dé Nguyen
- Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
46
|
Mehta S, Shelling A, Muthukaruppan A, Lasham A, Blenkiron C, Laking G, Print C. Predictive and prognostic molecular markers for cancer medicine. Ther Adv Med Oncol 2011; 2:125-48. [PMID: 21789130 DOI: 10.1177/1758834009360519] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the last 10 years there has been an explosion of information about the molecular biology of cancer. A challenge in oncology is to translate this information into advances in patient care. While there are well-formed routes for translating new molecular information into drug therapy, the routes for translating new information into sensitive and specific diagnostic, prognostic and predictive tests are still being developed. Similarly, the science of using tumor molecular profiles to select clinical trial participants or to optimize therapy for individual patients is still in its infancy. This review will summarize the current technologies for predicting treatment response and prognosis in cancer medicine, and outline what the future may hold. It will also highlight the potential importance of methods that can integrate molecular, histopathological and clinical information into a synergistic understanding of tumor progression. While these possibilities are without doubt exciting, significant challenges remain if we are to implement them with a strong evidence base in a widely available and cost-effective manner.
Collapse
Affiliation(s)
- Sunali Mehta
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
47
|
Robertson R, Germanos MS, Manfredi MG, Smith PG, Silva MD. Multimodal imaging with (18)F-FDG PET and Cerenkov luminescence imaging after MLN4924 treatment in a human lymphoma xenograft model. J Nucl Med 2011; 52:1764-9. [PMID: 21994410 DOI: 10.2967/jnumed.111.091710] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Cerenkov luminescence imaging (CLI) is an emerging imaging technique that combines aspects of both optical and nuclear imaging fields. The ability to fully evaluate the correlation and sensitivity of CLI to PET is critical to progress this technique further for use in high-throughput screening of pharmaceutical compounds. To achieve this milestone, it must first be established that CLI data correlate to PET data in an in vivo preclinical antitumor study. We used MLN4924, a phase 2 oncology therapeutic, which targets and inhibits the NEDD8-activating enzyme pathway involved in the ubiquitin-proteasome system. We compared the efficacious effects of MLN4924 using PET and Cerenkov luminescence image values in the same animals. METHODS Imaging of (18)F-FDG uptake was performed at 5 time points after drug treatment in the subcutaneously implanted diffuse large B-cell lymphoma tumor line OCI-Ly10. Data were acquired with both modalities on the same day, with a 15-min delay between CLI and PET. PET data analysis was performed using percentage injected dose per cubic centimeter of tissue (%ID/cm(3)), average standardized uptake values, and total glycolytic volume. CLI measurements were radiance, radiance per injected dose (radiance/ID), and total radiant volume. RESULTS A strong correlation was found between PET total glycolytic volume and CLI total radiant volume (r(2) = 0.99) and various PET and CLI analysis methods, with strong correlations found between PET %ID/cm(3) and CLI radiance (r(2) = 0.83) and CLI radiance/ID (r(2) = 0.82). MLN4924 demonstrated a significant reduction in tumor volume after treatment (volume ratio of treated vs. control, 0.114 at day 29). CONCLUSION The PET and CLI data presented confirm the correlation and dynamic sensitivity of this new imaging modality. CLI provides a preclinical alternative to expensive PET instrumentation. Future high-throughput studies should provide for quicker turnaround and higher cost-to-return benefits in the drug discovery process.
Collapse
Affiliation(s)
- Robbie Robertson
- Biomedical Imaging Group, Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, USA.
| | | | | | | | | |
Collapse
|
48
|
Non-invasive imaging in experimental medicine for drug development. Curr Opin Pharmacol 2011; 11:501-7. [DOI: 10.1016/j.coph.2011.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 04/17/2011] [Indexed: 01/01/2023]
|
49
|
Response to treatment series: part 2, tumor response assessment--using new and conventional criteria. AJR Am J Roentgenol 2011; 197:18-27. [PMID: 21701006 DOI: 10.2214/ajr.11.6581] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Conventional anatomic imaging biomarkers, including World Health Organization (WHO) criteria and Response Evaluation Criteria in Solid Tumors (RECIST), although effective, have limitations. This article will discuss the conventional and newer morphologic imaging biomarkers for the assessment of tumor response to therapy. CONCLUSION Applying established methods of assessing tumor response to therapy allows consistency in image interpretation and facilitates communication with oncologists. Because of the new methods of treatment, assessment of necrosis and volumetric information will need to be incorporated into size-based criteria.
Collapse
|
50
|
Wagner CC, Langer O. Approaches using molecular imaging technology -- use of PET in clinical microdose studies. Adv Drug Deliv Rev 2011; 63:539-46. [PMID: 20887762 DOI: 10.1016/j.addr.2010.09.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging uses minute amounts of radiolabeled drug tracers and thereby meets the criteria for clinical microdose studies. The advantage of PET, when compared to other analytical methods used in microdose studies, is that the pharmacokinetics (PK) of a drug can be determined in the tissue targeted for drug treatment. PET microdosing already offers interesting applications in clinical oncology and in the development of central nervous system pharmaceuticals and is extending its range of application to many other fields of pharmaceutical medicine. Although requirements for preclinical safety testing for microdose studies have been cut down by regulatory authorities, radiopharmaceuticals increasingly need to be produced under good manufacturing practice (GMP) conditions, which increases the costs of PET microdosing studies. Further challenges in PET microdosing include combining PET with other ultrasensitive analytical methods, such as accelerator mass spectrometry (AMS), to gain plasma PK data of drugs, beyond the short PET examination periods. Finally, conducting clinical PET studies with radiolabeled drugs both at micro- and therapeutic doses is encouraged to answer the question of dose linearity in clinical microdosing.
Collapse
Affiliation(s)
- Claudia C Wagner
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger-Gürtel 18-20, A-1090, Vienna, Austria
| | | |
Collapse
|